US5919456A - IL-13 receptor specific chimeric proteins - Google Patents

IL-13 receptor specific chimeric proteins Download PDF

Info

Publication number
US5919456A
US5919456A US08/821,840 US82184097A US5919456A US 5919456 A US5919456 A US 5919456A US 82184097 A US82184097 A US 82184097A US 5919456 A US5919456 A US 5919456A
Authority
US
United States
Prior art keywords
molecule
cells
receptor
chimeric
hil
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
US08/821,840
Inventor
Raj K. Puri
Waldemar Debinski
Ira Pastan
Nicholas Obiri
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
US Department of Health and Human Services
Original Assignee
US Department of Health and Human Services
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by US Department of Health and Human Services filed Critical US Department of Health and Human Services
Priority to US08/821,840 priority Critical patent/US5919456A/en
Application granted granted Critical
Publication of US5919456A publication Critical patent/US5919456A/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/642Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the peptide or protein in the drug conjugate being a cytokine, e.g. IL2, chemokine, growth factors or interferons being the inactive part of the conjugate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/21Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Pseudomonadaceae (F)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5437IL-13
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • This invention relates to methods of specifically delivering an effector molecule to a tumor cell.
  • this invention relates to chimeric molecules that specifically bind to IL-13 receptors and their use to deliver molecules having a particular activity to tumors overexpressing IL-13 receptors.
  • a chimeric molecule two or more molecules that exist separately in their native state are joined together to form a single molecule having the desired functionality of all of its constituent molecules.
  • one of the constituent molecules of a chimeric molecule is a "targeting molecule".
  • the targeting molecule is a molecule such as a ligand or an antibody that specifically binds to its corresponding target, for example a receptor on a cell surface.
  • the targeting molecule is an antibody
  • the chimeric molecule will specifically bind (target) cells and tissues bearing the epitope to which the antibody is directed.
  • the effector molecule refers to a molecule that is to be specifically transported to the target to which the chimeric molecule is specifically directed.
  • the effector molecule typically has a characteristic activity that is desired to be delivered to the target cell. Effector molecules include cytotoxins, labels, radionuclides, ligands, antibodies, drugs, liposomes, and the like.
  • first effector molecule is a cytotoxin
  • the chimeric molecule may act as a potent cell-killing agent specifically targeting the cytotoxin to cells bearing a particular target molecule.
  • chimeric fusion proteins which include interleukin 4 (IL-4) or transforming growth factor (TGF ⁇ ) fused to Pseudomonas exotoxin (PE) or interleukin 2 (IL-2) fused to Diphtheria toxin (DT) have been shown to specifically target and kill cancer cells (Pastan et al., Ann. Rev. Biochem., 61: 331-354 (1992)).
  • chimeric cytotoxins it is desirable to increase specificity and affinity and decrease cross-reactivity of the chimeric cytotoxins in order to increase their efficacy.
  • a target e.g. a tumor cell
  • a non-target e.g. a healthy cell
  • side effects of the chimeric molecule will be minimized.
  • targets to which chimeric molecules have been directed e.g. the IL-2 and IL-4 receptors
  • chimeric molecules directed to these targets show some adverse side-effects as they bind non-target cells that also express the targeted receptor.
  • the present invention provides methods and compositions for specifically delivering an effector molecule to a tumor cell.
  • the present invention provides chimeric molecules that specifically target tumor cells with less binding to healthy cells than other analogous chimeric molecules known in the prior art.
  • the improved specific targeting of this invention is premised, in part, on the discovery that tumor cells, especially carcinomas such as renal cell carcinoma, overexpress IL-13 receptors at extremely high levels.
  • the extremely high level of IL-13 receptor expression on target tumor cells permits the use of lower dosages of chimeric molecule to deliver the same amount of effector molecule to the target cells and also results in reduced binding of non-tumor cells.
  • this invention provides for a method for specifically delivering an effector molecule to a tumor cell bearing an IL-13 receptor.
  • the method involves providing a chimeric molecule comprising an effector molecule attached to a targeting molecule that specifically binds to an IL-13 receptor and contacting the tumor with the chimeric molecule resulting in binding of the chimeric molecule to the tumor cell.
  • the targeting molecule is preferably either a ligand, such as IL-13, that specifically binds an IL-13 receptor or an anti-IL-13 receptor antibody.
  • the targeting molecule may be conjugated to the effector molecule, or where both targeting and effector molecules are polypeptides, the targeting molecule may be joined to the effector molecule through one or more peptide bonds thereby forming a fusion protein.
  • Suitable effector molecules include a cytotoxin, a label, a radionuclide, a drug, a liposome, a ligand, and an antibody.
  • the effector is a cytotoxin, more specifically a Pseudomonas exotoxin such as PE38QQR. Where the Pseudomonas exotoxin is fused to an IL-13 targeting molecule, a preferred fusion protein is IL-13-PE38QQR.
  • this invention provides a method for impairing the growth of tumor cells, more preferably solid tumor cells, bearing an IL-13 receptor.
  • the method involves contacting the tumor with a chimeric molecule comprising an effector molecule selected from the group consisting of a cytotoxin, a radionuclide, a ligand and an antibody; said effector molecule being attached to a targeting molecule that specifically binds a human IL-13 receptor.
  • the targeting molecule is preferably a ligand (such as IL-13) that binds the IL-13 receptor or an anti-IL-13 receptor antibody.
  • Preferred cytotoxic effector molecules include Pseudomonas exotoxin, Diphtheria toxin, ricin and abrin.
  • Psuedomonas exotoxins such as PE38QQR
  • the targeting molecule may be conjugated or fused to the effector molecule with attachment by fusion preferred for cytotoxic effector molecules.
  • the tumor growth that is impaired may be tumor growth in a human.
  • the method may further comprise administering the chimeric molecule to a human intravenously into a body cavity, or into a human or an organ.
  • this invention provides for a method of detecting the presence or absence of a tumor.
  • the method involves contacting the tumor with a chimeric molecule comprising a detectable label attached to a targeting molecule that specifically binds a human IL-13 receptor and detecting the presence or absence of the label.
  • the label is selected from the group consisting of a radioactive label, an enzymatic label, an electron dense label, and a fluorescent label.
  • This invention also provides for vectors comprising a nucleic acid sequence encoding a chimeric polypeptide fusion protein comprising an IL-13 attached to a second polypeptide.
  • the chimeric polypeptide fusion protein specifically binds to a tumor cell bearing an IL-13 receptor.
  • a preferred vector encodes an IL-13-PE fusion protein and more preferably encodes an IL-13-PE38QQR fusion protein.
  • This invention also provides for host cells comprising a nucleic acid sequence encoding a chimeric polypeptide fusion protein comprising an IL-13 attached to a second polypeptide.
  • a preferred host cell comprises a nucleic acid encoding an IL-13-PE fusion protein, more preferably encoding an IL-13-PE38QQR fusion protein.
  • the encoded fusion protein specifically binds to a tumor cell bearing an IL-13 receptor.
  • Particularly preferred host cells are bacterial host cells, especially E. coli cells.
  • this invention provides chimeric molecules that specifically bind a tumor cell bearing an IL-13 receptor.
  • the chimeric molecule comprises a cytotoxic molecule attached to a targeting molecule that specifically binds an IL-13.
  • the targeting molecule may be conjugated or fused to the cytotoxic molecule.
  • the targeting molecule is fused to the cytotoxin thereby forming a single-chain fusion protein.
  • Particularly preferred targeting molecules are IL-13 or an antibody that specifically binds to the IL-13 receptor.
  • Preferred cytotoxic molecules include Pseudomonas exotoxin, Diphtheria toxin, ricin, and abrin, with Pseudomonas exotoxins (especially PE38QQR) being most preferred.
  • the chimeric molecule comprises an effector molecule attached to an antibody that specifically binds to an IL-13 receptor.
  • Effector molecules include a cytotoxin, a label, a radionuclide, a drug, liposome, a ligand and an antibody.
  • the effector molecule may be fused or conjugated to the antibody.
  • the invention additionally provides for pharmacological compositions comprising a pharmaceutically acceptable carrier and a chimeric molecule where the chimeric molecule comprises and effector molecule attached to a targeting molecule that specifically binds to an IL-13 receptor.
  • the targeting and effector molecules may be conjugated or fused to each other.
  • Particularly preferred targeting molecules include IL-13 and anti-IL-13 receptor antibodies, while preferred effector molecules include a cytotoxin, a label, a radionuclide, a drug, a liposome, a ligand and an antibody.
  • a preferred pharmacological composition includes an IL-13-PE fusion protein, more preferably an IL-13-PE38QQR fusion protein.
  • specifically deliver refers to the preferential association of a molecule with a cell or tissue bearing a particular target molecule or marker and not to cells or tissues lacking that target molecule. It is, of course, recognized that a certain degree of non-specific interaction may occur between a molecule and a non-target cell or tissue. Nevertheless, specific delivery, may be distinguished as mediated through specific recognition of the target molecule. Typically specific delivery results in a much stronger association between the delivered molecule and cells bearing the target molecule than between the delivered molecule and cells lacking the target molecule.
  • Specific delivery typically results in greater than 2 fold, preferably greater than 5 fold, more preferably greater than 10 fold and most preferably greater than 100 fold increase in amount of delivered molecule (per unit time) to a cell or tissue bearing the target molecule as compared to a cell or tissue lacking the target molecule or marker.
  • amino acid refers to an amino acid that is incorporated into a polypeptide.
  • the amino acid may be a naturally occurring amino acid and, unless otherwise limited, may encompass known analogs of natural amino acids that can function in a similar manner as naturally occurring amino acids.
  • a “fusion protein” refers to a polypeptide formed by the joining of two or more polypeptides through a peptide bond formed between the amino terminus of one polypeptide and the carboxyl terminus of another polypeptide.
  • the fusion protein may be formed by the chemical coupling of the constituent polypeptides or it may be expressed as a single polypeptide from nucleic acid sequence encoding the single contiguous fusion protein.
  • a single chain fusion protein is a fusion protein having a single contiguous polypeptide backbone.
  • a "spacer” as used herein refers to a peptide that joins the proteins comprising a fusion protein. Generally a spacer has no specific biological activity other than to join the proteins or to preserve some minimum distance or other spatial relationship between them. However, the constituent amino acids of a spacer may be selected to influence some property of the molecule such as the folding, net charge, or hydrophobicity of the molecule.
  • a “ligand”, as used herein, refers generally to all molecules capable of reacting with or otherwise recognizing or binding to a receptor on a target cell.
  • ligands include, but are not limited to, antibodies, lymphokines, cytokines, receptor proteins such as CD4 and CD8, solubilized receptor proteins such as soluble CD4, hormones, growth factors, and the like which specifically bind desired target cells.
  • the present invention provides a method for specifically delivering an effector molecule to a tumor cell.
  • This method involves the use of chimeric molecules comprising a targeting molecule attached to an effector molecule.
  • the chimeric molecules specifically target tumor cells while providing reduced binding to non-target cells as compared to other targeted chimeric molecules known in the art.
  • the improved specific targeting of this invention is premised, in part, on the discovery that solid tumors, especially carcinomas, overexpress IL-13 receptors at extremely high levels. While the IL-13 receptors are overexpressed on tumor cells, expression on other cells (e.g. monocytes and T cells) appears negligible. Thus, by specifically targeting the IL-13 receptor, the present invention provides chimeric molecules that are specifically directed to solid tumors while minimizing targeting of other cells or tissues.
  • this invention provides for compositions and methods for impairing the growth of tumors.
  • the methods involve providing a chimeric molecule comprising a cytotoxic effector molecule attached to a targeting molecule that specifically binds an IL-13 receptor.
  • the cytotoxin may be a native or modified cytotoxin such as Pseudomonas exotoxin (PE), Diphtheria toxin (DT), ricin, abrin, and the like.
  • the chimeric cytotoxin is administered to an organism containing tumor cells which are then contacted by the chimeric molecule.
  • the targeting molecule component of the chimeric molecule specifically binds to the overexpressed IL-13 receptors on the tumor cells. Once bound to the IL-13 receptor on the cell surface, the cytotoxic effector molecule mediates internalization into the cell where the cytotoxin inhibits cellular growth or kills the cell.
  • chimeric molecules comprising a targeting moiety joined to a cytotoxic effector molecules to target and kill tumor cells
  • chimeric fusion proteins which include interleukin 4 (IL-4) or transforming growth factor (TGF ⁇ ) fused to Pseudomonas exotoxin (PE) or interleukin 2 (IL-2) fused to Diphtheria toxin (DT) have been tested for their ability to specifically target and kill cancer cells (Pastan et al., Ann. Rev. Biochem., 61: 331-354 (1992)).
  • IL-4 shows some sequence similarity to IL-13
  • cytotoxins targeted by a moiety specific to the IL-13 receptor show significantly increased efficacy as compared to IL-4 receptor directed cytotoxins.
  • the improved efficacy of the IL-13 chimeras of the present invention is due to at least three factors.
  • IL-13 receptors are expressed at much lower levels, if at all on non-tumor cells (e.g. monocytes, T cells, B cells).
  • non-tumor cells e.g. monocytes, T cells, B cells.
  • cytotoxins directed to IL-13 receptors show reduced binding and subsequent killing of healthy cells and tissues as compared to cytotoxins directed to IL-4 receptors.
  • the receptor component that specifically binds IL-13 appears to be expressed at significantly higher levels on solid tumors than the receptor component that binds IL-4.
  • tumor cells bind higher levels of cytotoxic chimeric molecules directed against IL-13 receptors than cytotoxic chimeric molecules directed against IL-4 receptors.
  • IL-4 receptors are up-regulated when immune system cells (e.g. T-cells) are activated. This results in healthy cells, for example T-cells and B-cells, showing greater susceptibility to IL-4 receptor directed cytotoxins. Thus, the induction of an immune reaponse (as against a cancer), results in greater susceptiblity of cells of the immune system to the therapeutic agent.
  • IL-13 receptors are not up-regulated in activated cells. Thus IL-13 receptor targeted cytotoxins have no greater effect on activated cells and thereby minimize adverse effects of the therapeutic composition on cells of the immune system.
  • this invention also provides for compositions and methods for detecting the presence or absence of tumor cells. These methods involve providing a chimeric molecule comprising an effector molecule, that is a detectable label attached to a targeting molecule that specifically binds an IL-13 receptor.
  • the IL-13 receptor targeting moiety specifically binds the chimeric molecule to tumor cells which are then marked by their association with the detectable label. Subsequent detection of the cell-associated label indicates the presence of a tumor cell.
  • the effector molecule may be another specific binding moiety such as an antibody, a growth factor, or a ligand.
  • the chimeric molecule will then act as a highly specific bifunctional linker. This linker may act to bind and enhance the interaction between cells or cellular components to which the fusion protein binds.
  • the "targeting" component of the chimeric molecule comprises a polypeptide that specifically binds to an IL-13 receptor and the "effector" component is an antibody or antibody fragment (e.g. an Fv fragment of an antibody)
  • the targeting component specifically binds cancer cells, while the effector component binds receptors (e.g., IL-2 or IL-4 receptors) on the surface of immune cells.
  • the chimeric molecule may thus act to enhance and direct an immune response toward target cancer cells.
  • the effector molecule may be a pharmacological agent (e.g. a drug) or a vehicle containing a pharmacological agent.
  • a pharmacological agent e.g. a drug
  • vehicle containing a pharmacological agent e.g. a vehicle containing a pharmacological agent.
  • the moiety that specifically binds to an IL-13 receptor may be conjugated to a drug such as vinblastine, doxirubicin, genistein (a tyrosine kinase inhibitor), an antisense molecule, and other pharmacological agents known to those of skill in the art, thereby specifically targeting the pharmacological agent to tumor cells over expressing IL-13 receptors.
  • the targeting molecule may be bound to a vehicle containing the therapeutic composition.
  • vehicles include, but are not limited to liposomes, micelles, various synthetic beads, and the like.
  • the chimeric molecules of the present invention may include multiple targeting moieties bound to a single effector or conversely, multiple effector molecules bound to a single targeting moiety.
  • the chimeric molecules may include both multiple targeting moieties and multiple effector molecules.
  • this invention provides for "dual targeted" cytotoxic chimeric molecules in which targeting molecule that specifically binds to IL-13 is attached to a cytotoxic molecule and another molecule (e.g. an antibody, or another ligand) is attached to the other terminus of the toxin.
  • Such a dual-targeted cytotoxin might comprise an IL-13 substituted for domain Ia at the amino terminus of a PE and anti-TAC(Fv) inserted in domain III, between amino acid 604 and 609.
  • Other antibodies may also be suitable.
  • the targeting molecule is a molecule that specifically binds to the IL-13 receptor.
  • the specified ligand or antibody binds to its particular "target" protein (e.g. an IL-13 receptor protein) and does not bind in a significant amount to other proteins present in the sample or to other proteins to which the ligand or antibody may come in contact in an organism.
  • immunoassay formats may be used to select antibodies specifically immunoreactive with an IL-13 receptor protein.
  • solid-phase ELISA immunoassays are routinely used to select monoclonal antibodies specifically immunoreactive with a protein. See Harlow and Lane (1988) Antibodies, A Laboratory Manual, Cold Spring Harbor Publications, New York, for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity.
  • Example 1 provides a detailed protocol for assessing specific binding of labeled IL-13 by and IL-13 receptor.
  • the IL-13 receptor is a cell surface receptor that specifically binds IL-13 and mediates a variety of physiological responses in various cell types as described below in the description of IL-13.
  • the IL-13 receptor may be identified by contacting a cell or other sample with labeled IL-13 and detecting the amount of specific binding of IL-13 according to methods well known to those of skill in the art. Detection of IL-13 receptors by labeled IL-13 binding is described in detail in Example 1.
  • an anti-IL-13 receptor antibody may also be used to identify IL-13 receptors.
  • the antibody will specifically bind to the IL-13 receptor and this binding may be detected either through detection of a conjugated label or through detection of a labeled second antibody that binds the anti-IL-13 receptor antibody.
  • the moiety utilized to specifically target the IL-13 receptor is either an antibody that specifically binds the IL-13 receptor (an anti-IL-13R antibody) or a ligand, such as IL-13, that specifically binds to the receptor.
  • IL-13 is a pleiotropic cytokine that is recognized to share many of the properties of IL-4.
  • IL-13 has approximately 30% sequence identity with IL-4 and exhibits IL-4-like activities on monocytes/macrophages and human B cells (Minty et al., Nature, 362: 248 (1993), McKenzie et al. Proc. Natl. Acad. Sci. USA, 90: 3735 (1987)).
  • IL-13 appears to be a potent regulator of inflammatory and immune responses.
  • IL-13 can up-regulate the monocyte/macrophage expression of CD23 and MHC class I and class II antigens, down-regulate the expression of Fc ⁇ , and inhibit antibody-dependent cytotoxicity.
  • IL-13 can also inhibit nitric oxide production as well as the expression of pro-inflammatory cytokines (e.g. IL-1, IL-6, IL-8, IL-10 and IL-12) and chemokines (MIP-1, MCP), but enhance the production of IL-1ra (Minty supra.; Mckenzie et al., supra.; Zurawski et al. Immunol. Today, 15: 19 (1994); de Wall Malefyt et al. J.
  • pro-inflammatory cytokines e.g. IL-1, IL-6, IL-8, IL-10 and IL-12
  • MIP-1, MCP chemokines
  • Recombinant IL-13 is commercially available from a number of sources (see, e.g. R & D Systems, Minneapolis, Minn., USA, and Sanofi Bio-Industries, Inc., Tervose, Pa., USA).
  • a gene or a cDNA encoding IL-13 may be cloned into a plasmid or other expression vector and expressed in any of a number of expression systems according to methods well known to those of skill in the art. Methods of cloning and expressing IL-13 and the nucleic acid sequence for IL-13 are well kown (see, for example, Minty et al. (1993) supra. and McKenzie (1987), supra).
  • the expression of IL-13 as a component of a chimeric molecule is detailed in Example 4.
  • analogues or fragments of IL-13 bearing will also specifically bind to the IL-13 receptor.
  • conservative substitutions of residues e.g., a serine for an alanine or an aspartic acid for a glutamic acid
  • IL-13 when used in reference to a targeting molecule, also includes fragments, analogues or peptide mimetics of IL-13 that also specifically bind to the IL-13 receptor.
  • antibody includes various forms of modified or altered antibodies, such as an intact immunoglobulin, various fragments such as an Fv fragment, an Fv fragment containing only the light and heavy chain variable regions, an Fv fragment linked by a disulfide bond (Brinkmann, et al. Proc. Natl. Acad. Sci.
  • the antibody may be of animal (especially mouse or rat) or human origin or may be chimeric (Morrison et al., Proc Nat. Acad. Sci. USA 81: 6851-6855 (1984)) or humanized (Jones et al., Nature 321: 522-525 (1986), and published UK patent application #8707252).
  • Antibodies that specifically bind the IL-13 receptor may be produced by a number of means well known to those of skill in the art. Generally, this involves using an antigenic component of the IL-13 receptor as an antigen to induce the production of antibodies in an organism (e.g. a sheep, mouse, rabbit, etc.).
  • an antigenic component of the IL-13 receptor as an antigen to induce the production of antibodies in an organism (e.g. a sheep, mouse, rabbit, etc.).
  • an antigenic component of the IL-13 receptor as an antigen to induce the production of antibodies in an organism (e.g. a sheep, mouse, rabbit, etc.).
  • DSS disuccinimidyl suberate
  • the labeled receptor may then be isolated according to routine methods and the isolated receptor may be used as an antigen to raise anti-IL-13 receptor antibodies as described below.
  • Cross-linking and isolation of components of the IL-13 receptor is described in Example 3.
  • IL-13 receptors may be isolated by means of affinity chromatography. It was a surprising discovery of the present invention that solid tumor cells overexpress IL-13 receptors. This discovery of cells overexpressing IL-13 receptor greatly simplifies the receptor isolation. Generally, approximately, 100 million renal carcinoma cells, may be solubilized in detergent with protease inhibitors according to standard methods. The resulting lysate is then run through an affinity column bearing IL-13. The receptor binds to the IL-13 in the column thereby effecting an isolation from the lysate. The column is then eluted with a low pH buffer to dissociate the IL-13 ligand from the IL-13 receptor resulting in isolated receptor. The isolated receptor may then be used as an antigen to raise anti-IL-13 receptor antibodies.
  • an immunogen preferably an isolated IL-13 receptor or receptor epitope is mixed with an adjuvant and animals are immunized with the mixture.
  • the animal's immune response to the immunogen preparation is monitored by taking test bleeds and determining the titer of reactivity to the polypeptide of interest.
  • blood is collected from the animal and antisera are prepared. Further fractionation of the antisera to enrich for antibodies reactive to the polypeptide is performed where desired. See, e.g., Coligan (1991) Current Protocols in Immunology Wiley/Greene, New York; and Harlow and Lane (1989) Antibodies: A Laboratory Manual Cold Spring Harbor Press, New York, which are incorporated herein by reference.
  • Monoclonal antibodies may be obtained by various techniques familiar to those skilled in the art. Description of techniques for preparing such monoclonal antibodies may be found in, e.g., Stites et al. (eds.) Basic and Clinical Immunology (4th ed.) Lange Medical Publications, Los Altos, Calif., and references cited therein; Harlow and Lane (1988) Antibodies: A Laboratory Manual CSH Press; Goding (1986) Monoclonal Antibodies: Principles and Practice (2d ed.) Academic Press, New York, N.Y.; and particularly in Kohler and Milstein (1975) Nature 256: 495-497, which discusses one method of generating monoclonal antibodies.
  • this method involves injecting an animal with an immunogen. The animal is then sacrificed and cells taken from its spleen, which are then fused with myeloma cells (See, Kohler and Milstein (1976) Eur. J. Immunol. 6: 511-519, incorporated herein by reference). The result is a hybrid cell or "hybridoma" that is capable of reproducing in vitro.
  • Colonies arising from single immortalized cells are screened for production of antibodies of the desired specificity and affinity for the antigen, and yield of the monoclonal antibodies produced by such cells is enhanced by various techniques, including injection into the peritoneal cavity of a vertebrate host.
  • the individual antibody species obtained are the products of immortalized and cloned single B cells from the immune animal generated in response to a specific site recognized on the immunogenic substance.
  • suitable monoclonal antibodies will usually bind their target epitope with at least a K D of about 1 mM, more usually at least about 300 ⁇ M, and most preferably at least about 0.1 ⁇ M or better.
  • the chimeric molecule may contain targeting antibodies directed to tumor markers other than the overexpressed IL-13 receptor.
  • targeting antibodies directed to tumor markers other than the overexpressed IL-13 receptor.
  • a number of such antibodies are known and have even been converted to form suitable for incorporation into fusion proteins. These include anti-erbB2, B3, BR96, OVB3, anti-transferrin, Mik- ⁇ 1 and PR1 (see Batra et al., Mol. Cell. Biol., 11: 2200-2205 (1991); Batra et al., Proc. Natl. Acad. Sci. USA, 89: 5867-5871 (1992); Brinkmann, et al. Proc. Natl. Acad. Sci.
  • the effector molecule component of the chimeric molecules of this invention may be any molecule whose activity it is desired to deliver to cells that overexpress IL-13 receptors.
  • Particularly preferred effector molecules include cytotoxins such as PE or DT, radionuclides, ligands such as growth factors, antibodies, detectable labels such as fluorescent or radioactive labels, and therapeutic compositions such as liposomes and various drugs.
  • cytotoxins include Pseudomonas exotoxins, Diphtheria toxins, ricin, and abrin. Pseudomonas exotoxin and Dipthteria toxin are most preferred.
  • PE Pseudomonas exotoxin
  • Pseudomonas exotoxin A is an extremely active monomeric protein (molecular weight 66 kD), secreted by Pseudomonas aeruginosa, which inhibits protein synthesis in eukaryotic cells through the inactivation of elongation factor 2 (EF-2) by catalyzing its ADP-ribosylation (catalyzing the transfer of the ADP ribosyl moiety of oxidized NAD onto EF-2).
  • EF-2 elongation factor 2
  • the toxin contains three structural domains that act in concert to cause cytotoxicity.
  • Domain Ia (amino acids 1-252) mediates cell binding.
  • Domain II (amino acids 253-364) is responsible for translocation into the cytosol and domain III (amino acids 400-613) mediates ADP ribosylation of elongation factor 2, which inactivates the protein and causes cell death.
  • domain Ib (amino acids 365-399) remains undefined, although a large part of it, amino acids 365-380, can be deleted without loss of cytotoxicity. See Siegall et al., J. Biol. Chem. 264: 14256-14261 (1989), incorporated by reference herein.
  • a preferred PE molecule is one in which domain Ia (amino acids 1 through 252) is deleted and amino acids 365 to 380 have been deleted from domain Ib.
  • domain Ia amino acids 1 through 252
  • amino acids 365 to 380 have been deleted from domain Ib.
  • all of domain Ib and a portion of domain II can be deleted, particularly if the deleted sequences are replaced with a linking peptide such as GGGGS (SEQ ID NO:1).
  • PE molecules can be further modified using site-directed mutagenesis or other techniques known in the art, to alter the molecule for a particular desired application.
  • Means to alter the PE molecule in a manner that does not substantially affect the functional advantages provided by the PE molecules described here can also be used and such resulting molecules are intended to be covered herein.
  • REDLK SEQ ID NO:2
  • REDL SEQ ID NO:3
  • RDEL SEQ ID NO:4
  • KDEL SEQ ID NO:5
  • Deletions of amino acids 365-380 of domain Ib can be made without loss of activity. Further, a substitution of methionine at amino acid position 280 in place of glycine to allow the synthesis of the protein to begin and of serine at amino acid position 287 in place of cysteine to prevent formation of improper disulfide bonds is beneficial.
  • the targeting molecule is inserted in replacement for domain Ia. A similar insertion has been accomplished in what is known as the TGF ⁇ -PE40 molecule (also referred to as TP40) described in Heimbrook et al., Proc. Natl. Acad. Sci., USA, 87: 4697-4701 (1990) and in commonly assigned U.S. Ser. No. 07/865,722 filed Apr. 8, 1992 and in U.S. Ser. No. 07/522,563 filed May 14, 1990, all of which are incorporated by reference.
  • PE contains amino acids 253-364 and 381-608, and are followed by the native sequences REDLK (SEQ ID NO:2) or the mutant sequences KDEL (SEQ ID NO:5) or RDEL (SEQ ID NO:4).
  • Lysines at positions 590 and 606 may or may not be mutated to glutamine.
  • the IL-13 receptor targeted cytotoxins of this invention comprise the PE molecule designated PE38QQR.
  • This PE molecule is a truncated form of PE composed of amino acids 253-364 and 381-608.
  • the lysine residues at positions 509 and 606 are replaced by glutamine and at 613 are replaced by arginine (Debinski et al. Bioconj. Chem., 5: 40 (1994) which is incorporated herein by reference).
  • the targeting molecule (e.g. IL-13 or anti-IL-13R antibody) may also be inserted at a point within domain III of the PE molecule. Most preferably the targeting molecule is fused between about amino acid positions 607 and 609 of the PE molecule. This means that the targeting molecule is inserted after about amino acid 607 of the molecule and an appropriate carboxyl end of PE is recreated by placing amino acids about 604-613 of PE after the targeting molecule. Thus, the targeting molecule is inserted within the recombinant PE molecule after about amino acid 607 and is followed by amino acids 604-613 of domain III. The targeting molecule may also be inserted into domain Ib to replace sequences not necessary for toxicity. Debinski, et al. Mol. Cell. Biol., 11: 1751-1753 (1991).
  • the PE molecules will be fused to the targeting molecule by recombinant means.
  • the genes encoding protein chains may be cloned in cDNA or in genomic form by any cloning procedure known to those skilled in the art. See for example Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, (1989), incorporated by reference herein. Methods of cloning genes encoding PE fused to various ligands are well known to those of skill in the art. See, for example, Siegall et al., FASEB J., 3: 2647-2652 (1989); Chaudhary et al. Proc. Natl. Acad. Sci. USA, 84: 4538-4542 (1987), which are incorporated herein by reference.
  • deletions or changes may be made in PE or in a linker connecting an antibody gene to PE, in order to increase cytotoxicity of the fusion protein toward target cells or to decrease nonspecific cytotoxicity toward cells without antigen for the antibody. All such constructions may be made by methods of genetic engineering well known to those skilled in the art (see, generally, Sambrook et al., supra) and may produce proteins that have differing properties of affinity, specificity, stability and toxicity that make them particularly suitable for various clinical or biological applications.
  • diphtheria toxin kills cells by ADP-ribosylating elongation factor 2 thereby inhibiting protein synthesis. Diphtheria toxin, however, is divided into two chains, A and B, linked by a disulfide bridge. In contrast to PE, chain B of DT, which is on the carboxyl end, is responsible for receptor binding and chain A, which is present on the amino end, contains the enzymatic activity (Uchida et al., Science, 175: 901-903 (1972); Uchida et al. J. Biol. Chem., 248: 3838-3844 (1973)).
  • the targeting molecule-Diphtheria toxin fusion proteins of this invention have the native receptor-binding domain removed by truncation of the Diphtheria toxin B chain.
  • Particularly preferred is DT388, a DT in which the carboxyl terminal sequence beginning at residue 389 is removed.
  • the DT molecules may be chemically conjugated to the IL-13 receptor targeting molecule, but, in a preferred embodiment, the targeting molecule will be fused to the Diphtheria toxin by recombinant means.
  • the genes encoding protein chains may be cloned in cDNA or in genomic form by any cloning procedure known to those skilled in the art. Methods of cloning genes encoding DT fused to various ligands are also well known to those of skill in the art. See, for example, Williams et al. J. Biol. Chem. 265: 11885-11889 (1990) and copending patent application (U.S. Ser. No. 07/620,939) which describe the expression of a number of growth-factor-DT fusion proteins.
  • DT diphtheria toxin
  • Modifications typically include removal of the targeting domain in the B chain and, more specifically, involve truncations of the carboxyl region of the B chain.
  • Detectable labels suitable for use as the effector molecule component of the chimeric molecules of this invention include any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means.
  • Useful labels in the present invention include magnetic beads (e.g.
  • DYNABEADSTM fluorescent dyes (e.g., fluorescein isothiocyanate, texas red, rhodamine, green fluorescent protein, and the like), radiolabels (e.g., 3 H, 125 I, 35 S, 14 C, or 32P), enzymes (e.g., horse radish peroxidase, alkaline phosphatase and others commonly used in an ELISA), and colorimetric labels such as colloidal gold or colored glass or plastic (e.g. polystyrene, polypropylene, latex, etc.) beads.
  • fluorescent dyes e.g., fluorescein isothiocyanate, texas red, rhodamine, green fluorescent protein, and the like
  • radiolabels e.g., 3 H, 125 I, 35 S, 14 C, or 32P
  • enzymes e.g., horse radish peroxidase, alkaline phosphatase and others commonly used in
  • radiolabels may be detected using photographic film or scintillation counters
  • fluorescent markers may be detected using a photodetector to detect emitted illumination
  • Enzymatic labels are typically detected by providing the enzyme with a substrate and detecting the reaction product produced by the action of the enzyme on the substrate, and colorimetric labels are detected by simply visualizing the colored label.
  • the effector molecule may also be a ligand or an antibody.
  • Particularly preferred ligand and antibodies are those that bind to surface markers on immune cells.
  • Chimeric molecules utilizing such antibodies as effector molecules act as bifunctional linkers establishing an association between the immune cells bearing binding partner for the ligand or antibody and the tumor cells overexpressing the IL-13 receptor.
  • Suitable antibodies and growth factors are known to those of skill in the art and include, but are not limited to, IL-2, IL-4, IL-6, IL-7, tumor necrosis factor (TNF), anti-Tac, TGF ⁇ , and the like.
  • Suitable effector molecules include pharmacological agents or encapsulation systems containing various pharmacological agents.
  • the targeting molecule of the chimeric molecule may be attached directly to a drug that is to be delivered directly to the tumor.
  • drugs are well known to those of skill in the art and include, but are not limited to, doxirubicin, vinblastine, genistein, an antisense molecule, and the like.
  • the effector molecule may be an encapsulation system, such as a liposome or micelle that contains a therapeutic composition such as a drug, a nucleic acid (e.g. an antisense nucleic acid), or another therapeutic moiety that is preferably shielded from direct exposure to the circulatory system.
  • a therapeutic composition such as a drug, a nucleic acid (e.g. an antisense nucleic acid), or another therapeutic moiety that is preferably shielded from direct exposure to the circulatory system.
  • a nucleic acid e.g. an antisense nucleic acid
  • another therapeutic moiety that is preferably shielded from direct exposure to the circulatory system.
  • Means of preparing liposomes attached to antibodies are well known to those of skill in the art. See, for example, U.S. Pat. No. 4,957,735, Connor et al., Pharm. Ther., 28: 341-365 (1985).
  • the targeting molecule and effector molecules may be joined together in any order.
  • the effector molecule may be joined to either the amino or carboxy termini of the targeting molecule.
  • the targeting molecule may also be joined to an internal region of the effector molecule, or conversely, the effector molecule may be joined to an internal location of the targeting molecule, as long as the attachment does not interfere with the respective activities of the molecules.
  • the targeting molecule and the effector molecule may be attached by any of a number of means well known to those of skill in the art.
  • the effector molecule is conjugated, either directly or through a linker (spacer), to the targeting molecule.
  • linker spacer
  • both the effector molecule and the targeting molecule are polypeptides it is preferable to recombinantly express the chimeric molecule as a single-chain fusion protein.
  • the targeting molecule e.g. IL-13 or anti-IL-13R antibody
  • the effector molecule e.g. a cytotoxin, a label, a ligand, or a drug or liposome.
  • Means of chemically conjugating molecules are well known to those of skill.
  • polypeptides typically contain variety of functional groups; e.g., carboxylic acid (COOH) or free amine (--NH 2 ) groups, which are available for reaction with a suitable functional group on an effector molecule to bind the effector thereto.
  • functional groups e.g., carboxylic acid (COOH) or free amine (--NH 2 ) groups, which are available for reaction with a suitable functional group on an effector molecule to bind the effector thereto.
  • the targeting molecule and/or effector molecule may be derivatized to expose or attach additional reactive functional groups.
  • the derivatization may involve attachment of any of a number of linker molecules such as those available from Pierce Chemical Company, Rockford Ill.
  • a “linker”, as used herein, is a molecule that is used to join the targeting molecule to the effector molecule.
  • the linker is capable of forming covalent bonds to both the targeting molecule and to the effector molecule.
  • Suitable linkers are well known to those of skill in the art and include, but are not limited to, straight or branched-chain carbon linkers, heterocyclic carbon linkers, or peptide linkers.
  • the linkers may be joined to the constituent amino acids through their side groups (e.g., through a disulfide linkage to cysteine). However, in a preferred embodiment, the linkers will be joined to the alpha carbon amino and carboxyl groups of the terminal amino acids.
  • a bifunctional linker having one functional group reactive with a group on a particular agent, and another group reactive with an antibody may be used to form the desired immunoconjugate.
  • derivatization may involve chemical treatment of the targeting molecule, e.g., glycol cleavage of the sugar moiety of a the glycoprotein antibody with periodate to generate free aldehyde groups.
  • the free aldehyde groups on the antibody may be reacted with free amine or hydrazine groups on an agent to bind the agent thereto.
  • Procedures for generation of free sulfhydryl groups on polypeptide, such as antibodies or antibody fragments are also known (See U.S. Pat. No. 4,659,839).
  • chimeric conjugates comprising linkages which are cleavable in the vicinity of the target site may be used when the effector is to be released at the target site. Cleaving of the linkage to release the agent from the antibody may be prompted by enzymatic activity or conditions to which the immunoconjugate is subjected either inside the target cell or in the vicinity of the target site.
  • a linker which is cleavable under conditions present at the tumor site e.g. when exposed to tumor-associated enzymes or acidic pH
  • cleavable linkers are known to those of skill in the art. See U.S. Pat. Nos. 4,618,492; 4,542,225, and 4,625,014.
  • the mechanisms for release of an agent from these linker groups include, for example, irradiation of a photolabile bond and acid-catalyzed hydrolysis.
  • U.S. Pat. No. 4,671,958, for example includes a description of immunoconjugates comprising linkers which are cleaved at the target site in vivo by the proteolytic enzymes of the patient's complement system.
  • the targeting molecule and/or the effector molecule is relatively short (i.e., less than about 50 amino acids) they may be synthesized using standard chemical peptide synthesis techniques. Where both molecules are relatively short the chimeric molecule may be synthesized as a single contiguous polypeptide. Alternatively the targeting molecule and the effector molecule may be synthesized separately and then fused by condensation of the amino terminus of one molecule with the carboxyl terminus of the other molecule thereby forming a peptide bond. Alternatively, the targeting and effector molecules may each be condensed with one end of a peptide spacer molecule thereby forming a contiguous fusion protein.
  • Solid phase synthesis in which the C-terminal amino acid of the sequence is attached to an insoluble support followed by sequential addition of the remaining amino acids in the sequence is the preferred method for the chemical synthesis of the polypeptides of this invention.
  • Techniques for solid phase synthesis are described by Barany and Merrifield, Solid-Phase Peptide Synthesis; pp. 3-284 in The Peptides: Analysis, Synthesis, Biology. Vol. 2: Special Methods in Peptide Synthesis, Part A., Merrifield, et al. J. Am. Chem. Soc., 85: 2149-2156 (1963), and Stewart et al., Solid Phase Peptide Synthesis, 2nd ed. Pierce Chem. Co., Rockford, Ill. (1984) which are incorporated herein by reference.
  • the chimeric fusion proteins of the present invention are synthesized using recombinant DNA methodology. Generally this involves creating a DNA sequence that encodes the fusion protein, placing the DNA in an expression cassette under the control of a particular promoter, expressing the protein in a host, isolating the expressed protein and, if required, renaturing the protein.
  • DNA encoding the fusion proteins (e.g. IL-13-PE38QQR) of this invention may be prepared by any suitable method, including, for example, cloning and restriction of appropriate sequences or direct chemical synthesis by methods such as the phosphotriester method of Narang et al. Meth. Enzymol. 68: 90-99 (1979); the phosphodiester method of Brown et al., Meth. Enzymol. 68: 109-151 (1979); the diethylphosphoramidite method of Beaucage et al., Tetra. Lett., 22: 1859-1862 (1981); and the solid support method of U.S. Pat. No. 4,458,066, all incorporated by reference herein.
  • Chemical synthesis produces a single stranded oligonucleotide. This may be converted into double stranded DNA by hybridization with a complementary sequence, or by polymerization with a DNA polymerase using the single strand as a template.
  • a complementary sequence or by polymerization with a DNA polymerase using the single strand as a template.
  • One of skill would recognize that while chemical synthesis of DNA is limited to sequences of about 100 bases, longer sequences may be obtained by the ligation of shorter sequences.
  • subsequences may be cloned and the appropriate subsequences cleaved using appropriate restriction enzymes. The fragments may then be ligated to produce the desired DNA sequence.
  • DNA encoding fusion proteins of the present invention may be cloned using DNA amplification methods such as polymerase chain reaction (PCR).
  • PCR polymerase chain reaction
  • the gene for IL-13 is PCR amplified, using a sense primer containing the restriction site for NdeI and an antisense primer containing the restriction site for HindIII.
  • the primers are selected to amplify the nucleic acid starting at position 19, as described by McKenzie et al. (1987), supra. This produces a nucleic acid encoding the mature IL-13 sequence and having terminal restriction sites.
  • a PE38QQR fragment may be cut out of the plasmid pWDMH4-38QQR or plasmid pSGC242FdN1 described by Debinski et al. Int. J. Cancer, 58: 744-748 (1994), and by Debinski et al. Clin. Res., 42: 251A (abstract (1994) respectively.
  • Ligation of the IL-13 and PE38QQR sequences and insertion into a vector produces a vector encoding IL-13 joined to the amino terminus of PE38QQR (position 253 of PE).
  • the two molecues are joined by a three amino acid junction consaisting of glutamic acid, alanine, and phenylalanine introduced by the restriction site.
  • the two molecules are preferrably essentially directly joined together, one of skill will appreciate that the molecules may be separated by a peptide spacer consisting of one or more amino acids.
  • the spacer will have no specific biological activity other than to join the proteins or to preserve some minimum distance or other spatial relationship between them.
  • the constituent amino acids of the spacer may be selected to influence some property of the molecule such as the folding, net charge, or hydrophobicity.
  • the nucleic acid sequences encoding the fusion proteins may be expressed in a variety of host cells, including E. coli, other bacterial hosts, yeast, and various higher eukaryotic cells such as the COS, CHO and HeLa cells lines and myeloma cell lines.
  • the recombinant protein gene will be operably linked to appropriate expression control sequences for each host.
  • this includes a promoter such as the T7, trp, or lambda promoters, a ribosome binding site and preferably a transcription termination signal.
  • control sequences will include a promoter and preferably an enhancer derived from immunoglobulin genes, SV40, cytomegalovirus, etc., and a polyadenylation sequence, and may include splice donor and acceptor sequences.
  • the plasmids of the invention can be transferred into the chosen host cell by well-known methods such as calcium chloride transformation for E. coli and calcium phosphate treatment or electroporation for mammalian cells.
  • Cells transformed by the plasmids can be selected by resistance to antibiotics conferred by genes contained on the plasmids, such as the amp, gpt, neo and hyg genes.
  • the recombinant fusion proteins can be purified according to standard procedures of the art, including ammonium sulfate precipitation, affinity columns, column chromatography, gel electrophoresis and the like (see, generally, R. Scopes, Protein Purification, Springer-Verlag, New York (1982), Guider, Methods in Enzymology Vol. 182: Guide to Protein Purification, Academic Press, Inc. New York (1990)). Substantially pure compositions of at least about 90 to 95% homogeneity are preferred, and 98 to 99% or more homogeneity are most preferred for pharmaceutical uses. Once purified, partially or to homogeneity as desired, the polypeptides may then be used therapeutically.
  • the IL-13 receptor targeted fusion protein may possess a conformation substantially different than the native conformations of the constituent polypeptides. In this case, it may be necessary to denature and reduce the polypeptide and then to cause the polypeptide to re-fold into the preferred conformation. Methods of reducing and denaturing proteins and inducing re-folding are well known to those of skill in the art. (See, Debinski et al. J. Biol. Chem., 268: 14065-14070 (1993); Kreitman and Pastan, Bioconjug. Chem., 4: 581-585 (1993); and Buchner, et al., Anal.
  • Debinski et al. describe the denaturation and reduction of inclusion body proteins in guanidine-DTE. The protein is then refolded in a redox buffer containing oxidized glutathione and L-arginine.
  • modifications can be made to the IL-13 receptor targeted fusion proteins without diminishing their biological activity. Some modifications may be made to facilitate the cloning, expression, or incorporation of the targeting molecule into a fusion protein. Such modifications are well known to those of skill in the art and include, for example, a methionine added at the amino terminus to provide an initiation site, or additional amino acids placed on either terminus to create conveniently located restriction sites or termination codons.
  • carcinoma tumor cells e.g. renal carcinoma cells
  • IL-13 receptors at levels ranging from about 2100 sites/cell to greater than 150,000 sites per cell.
  • IL-13 receptor IL-13 receptor
  • identification of other cells that overexpress IL-13 receptors requires only routine screening using well-known methods. Typically this involves providing a labeled molecule that specifically binds to the IL-13 receptor. The cells in question are then contacted with this molecule and washed. Quantification of the amount of label remaining associated with the test cell provides a measure of the amount of IL-13 receptor (IL-13R) present on the surface of that cell.
  • IL-13R IL-13 receptor
  • IL-13 receptor may be quantified by measuring the binding of 125 I-labeled IL-13 ( 125 I-IL-13) to the cell in question. Details of such a binding assay are provided in Example 1.
  • the chimeric molecules of this invention are useful for parenteral, topical, oral, or local administration, such as by aerosol or transdermally, for prophylactic and/or therapeutic treatment.
  • the pharmaceutical compositions can be administered in a variety of unit dosage forms depending upon the method of administration.
  • unit dosage forms suitable for oral administration include powder, tablets, pills, capsules and lozenges.
  • the fusion proteins and pharmaceutical compositions of this invention when administered orally, must be protected from digestion. This is typically accomplished either by complexing the protein with a composition to render it resistant to acidic and enzymatic hydrolysis or by packaging the protein in an appropriately resistant carrier such as a liposome. Means of protecting proteins from digestion are well known in the art.
  • compositions of this invention are particularly useful for parenteral administration, such as intravenous administration or administration into a body cavity or lumen of an organ.
  • the compositions for administration will commonly comprise a solution of the chimeric molecule dissolved in a pharmaceutically acceptable carrier, preferably an aqueous carrier.
  • a pharmaceutically acceptable carrier preferably an aqueous carrier.
  • aqueous carriers can be used, e.g., buffered saline and the like. These solutions are sterile and generally free of undesirable matter.
  • These compositions may be sterilized by conventional, well known sterilization techniques.
  • compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, toxicity adjusting agents and the like, for example, sodium acetate, sodium chloride, potassium chloride, calcium chloride, sodium lactate and the like.
  • concentration of chimeric molecule in these formulations can vary widely, and will be selected primarily based on fluid volumes, viscosities, body weight and the like in accordance with the particular mode of administration selected and the patient's needs.
  • a typical pharmaceutical composition for intravenous administration would be about 0.1 to 10 mg per patient per day. Dosages from 0.1 up to about 100 mg per patient per day may be used, particularly when the drug is administered to a secluded site and not into the blood stream, such as into a body cavity or into a lumen of an organ. Actual methods for preparing parenterally administrable compositions will be known or apparent to those skilled in the art and are described in more detail in such publications as Remington's Pharmaceutical Science, 15th ed., Mack Publishing Company, Easton, Pa. (1980).
  • compositions containing the present fusion proteins or a cocktail thereof can be administered for therapeutic treatments.
  • compositions are administered to a patient suffering from a disease, in an amount sufficient to cure or at least partially arrest the disease and its complications.
  • An amount adequate to accomplish this is defined as a "therapeutically effective dose.” Amounts effective for this use will depend upon the severity of the disease and the general state of the patient's health.
  • compositions may be administered depending on the dosage and frequency as required and tolerated by the patient.
  • the composition should provide a sufficient quantity of the proteins of this invention to effectively treat the patient.
  • cytotoxic fusion proteins of the present invention are included a variety of disease conditions caused by specific human cells that may be eliminated by the toxic action of the protein.
  • One preferred application is the treatment of cancer, such as by the use of an IL-13 receptor targeting molecule (e.g. IL-13 or anti-IL-13R antibody) attached to a cytotoxin.
  • an IL-13 receptor targeting molecule e.g. IL-13 or anti-IL-13R antibody
  • ligand portion of the molecule is chosen according to the intended use.
  • Proteins on the membranes of T cells that may serve as targets for the ligand includes CD2 (T11), CD3, CD4 and CD8. Proteins found predominantly on B cells that might serve as targets include CD10 (CALLA antigen), CD19 and CD20.
  • CD45 is a possible target that occurs broadly on lymphoid cells. These and other possible target lymphocyte target molecules for the chimeric molecules bearing a ligand effector are described in Leukocyte Typing III, A. J. McMichael, ed., Oxford University Press (1987). Those skilled in the art will realize ligand effectors may be chosen that bind to receptors expressed on still other types of cells as described above, for example, membrane glycoproteins or ligand or hormone receptors such as epidermal growth factor receptor and the like.
  • kits for the treatment of tumors or for the detection of cells overexpressing IL-13 receptors will typically comprise a chimeric molecule of the present invention (e.g. IL-13-label, IL-13-cytotoxin, IL-13-ligand, etc.).
  • the kits will typically include instructional materials disclosing means of use of chimeric molecule (e.g. as a cytotoxin, for detection of tumor cells, to augment an immune response, etc.).
  • the kits may also include additional components to facilitate the particular application for which the kit is designed.
  • the kit may additionally contain means of detecting the label (e.g.
  • kits may additionally include buffers and other reagents routinely used for the practice of a particular method. Such kits and appropriate contents are well known to those of skill in the art.
  • IL-4 and IL-13 were labeled with 125 I (Amersham Research Products, Arlington Heights, Ill., USA) by using the IODO-GEN reagent (Pierce, Rockford, Ill., USA) according to the manufacturer's instructions.
  • the specific activity of the radiolabeled cytokines was estimated to range from 20-100 ⁇ Ci/ ⁇ g protein.
  • RRC renal cell carcinoma
  • IL-13R expression was examined as previously described (Obiri et al. J.
  • IL-13 responsiveness has previously been reported in human monocytes, B cells and pre-myeloid (TF-1) cells (see, e.g. de Waal Malefyt, et al. J. Immunol., 151: 6370-6381 (1993), de Waal Malefyt, et al. J. Immunol., 144: 629-633 (1993)), little was known about IL-13R structure or its binding characteristics in these, or any other cells.
  • the present data show that freshly isolated human monocytes, EBV-transformed B cell line and TF-1 cell line express very few IL-13 binding sites (100-300/cell) compared to human RCC cells (Table 1).
  • IL-2R ⁇ c receptor subunit is associated with IL-13R (see, e.g., Russell et al. Science 262: 1880-1883 (1993); Kondo et al. Science, 262: 1874-1877 (1993); Noguchi et al. Science, 262: 1877-1880 (1993); Kondo et al. Science 263: 1453-1454 (1994); Giri et al. EMBO J. 13: 2822-2830 (1994))) and IL-13R may share a common component with IL-4R (Zurawski et al. EMBO J. 12: 2663-2670 (1993); Aversa et al. J. Exp. Med.
  • Unlabeled IL-4 more efficiently inhibited 125 I-IL-4 from binding to RCC cells (84%, and 72% displacement of total binding in WS-RCC and HL-RCC, respectively) than IL-13 which also displaced 125 I-IL-4 binding to these cells (61% of total binding in WS-RCC and 51% in HL-RCC) under similar conditions.
  • IL-13 binding was effectively displaced by IL-13 (about 85% of total in both cell types), it was only minimally displaced by IL-4 (12% of total displacement in WS-RCC, and 7% in HL-RCC).
  • lymphoid MLA 144 cells and RAJI cell lines were incubated with radiolabled IL-4 with or without excess unlabeled IL-4 or IL-13. Excess unlabeled IL-4 effectively displaced labeled 125 I-IL-4 bound to MLA 144 and RAJI cells, while excess IL-13 could not compete this binding. This observation is at variance to that seen with RCC cells in which IL-13 competed for IL-4 binding. The inability of IL-13 to compete for 125 I-IL-4 binding to MLA 144 is consistent with the observation that IL-13 did not bind to peripheral blood T (or MLA 144) cells.
  • the subunit structure of IL-13R on RCC cells was investigated by crosslinking studies.
  • Cells (5 ⁇ 10 6 ) were labeled with 125 I-IL-13 or 125 I-IL-4 in the presence or absence of excess IL-13 or IL-4 for 2 h at 4° C.
  • the bound ligand was cross-linked to its receptor with disuccinimidyl suberate (DSS) (Pierce, Rockford, Ill., USA) at a final concentration of 2 mM for 30 min.
  • DSS disuccinimidyl suberate
  • Cells were lysed in a buffer containing 1% Triton X-100, 1 mM phenylmethylsulfonyl fluoride, 0.02 mM leupeptin, 5.0 ⁇ M trypsin inhibitor, 10 mM benzamidine HCl, 1 mM phenanthroline iodoacetamide, 50 mM amino caproic acid, 10 ⁇ g/ml pepstatin, and 10 ⁇ g/ml aprotinin.
  • the cell lysates were cleared by boiling in buffer containing 2-mercaptoethanol and analyzed by electrophoresis through 8% SDS/polyacrylamide gel. The gel was subsequently dried and autoradiographed.
  • the receptor/ligand complex was immunoprecipitated from the lysate overnight at 4° C. by incubating with protein A sepharose beads that had been pre-incubated with P7 anti hIL-4R or anti- ⁇ c antibody and analyzed as above.
  • This band appeared to be specifically associated with IL-13R because unlabeled IL-13 competed for the binding of 125 I-IL-13. This band could represent an IL-13R associated protein or a proteolytic fragment of the larger band. In contrast to the displacement of 125 I-IL-13 binding by unlabeled IL-13, an excess of unlabeled IL-4 did not prevent the appearance of IL-13R band in RCC cell lines. IL-13 on the other hand competed for 125 I-IL-4 binding to both major proteins on WS-RCC cells.
  • 125 I-IL-13-cross-linked protein was slightly larger in size in TF-1.J61, WS-RCC, PM-RCC, and HL-RCC cell lines compared to that seen in MA-RCC. Post-translational modifications, such as glycosylation or phosphorylation, may account for this difference.
  • a coding region of the human interleukin 13 (hIL-13) gene (plasmid JFE14-SR ⁇ ) (Minty et al., Nature, 362: 248 (1993), McKenzie et al. Proc. Natl. Acad. Sci. USA, 90: 3735 (1987) which are incorporated herein by reference) was fused to a gene encoding PE38QQR, a mutated form of PE, thereby producing a construct (phuIL-13-Tx) encoding the chimeric molecule.
  • a DNA encoding human IL-13 was PCR-amplified from plasmid JFE14-SR ⁇ .
  • New sites were introduced for the restriction endonucleases NdeI and Hind III at the 5' and 3' ends of the hIL-13 gene, respectively by PCR using a sense primer that incorporated the NdeI site and an antisense primer that incorporated the HindIII site.
  • the NdeII/HindIII fragment containing encoding hIL-13 was subcloned into a vector obtained by digestion of plasmid pWDMH4-38QQR (Debinski et al. Int. J. Cancer 58: 744-748 (1994)) or plasmid pSGC242FdN1 (Debinski et al. Clin. Res. 42: 251A, (abstr.) (1994) with NdeI and HindIII, to produce plasmid phuIL-13-Tx.
  • the 5' end of the gene fusion was sequenced and showed the correct DNA of hIL-13.
  • hIL-4 Human interleukin 4
  • plasmid pWDMH4 (Debinski et al. J. Biol. Chem. 268: 14065-14070 (1993)) as a template for PCR amplification.
  • Recombinant proteins were expressed in E. coli BL21 ( ⁇ DE3) under control of the T7 late promoter (Id.).
  • the plasmids also carried a T7 transcription terminator at the end of the open reading frame of the protein, an f1 origin of replication and gene for ampicillin resistance (Debinski et al. J. Clin. Invest. 90: 405-411 (1992)).
  • the plasmids were amplified in E. coli (HB101 or DH5 ⁇ high efficiency transformation) (BRL) and DNA was extracted using Qiagen kits (Chatsworth, Calif., USA).
  • E. coli BL21 ( ⁇ DE3) cells were transformed with plasmids of interest and cultured in 1.0 liter of Super broth.
  • Expressed recombinant human IL-13 and human IL-13-PE38QQR were localized in inclusion bodies.
  • the recombinant proteins were isolated from the inclusion bodies as described by Debinski et al., J. Biol. Chem. 268: 14065-14070 (1993), which is incorporated herein by reference.
  • the renatured protein of human IL-13-PE38QQR was purified on Q-Sepharose Fast Flow and by size exclusion chromatography on Sephacryl S-200HR (Pharmacia, Piscataway, N.J., USA)
  • the initial step of hIL-13 or hIL-4 purification was conducted on SP-Sepharose Fast Flow (Pharmacia).
  • Protein concentration was determined by the Bradford assay (Pierce “Plus”, Rockford, Ill. USA) using BSA as a standard.
  • Human IL-13 and IL-13-PE38QQR were expressed at high levels in bacteria as seen in SDS-PAGE analysis of the total cell extract. After initial purification on SP-Sepharose (hIL-13) or Q-Sepharose (hIL-13-PE38QQR) the renatured recombinant proteins were applied onto a Sephacryl S-200 HR Pharmacia column. Human IL-13 and hIL-13-PE38QQR appeared as single entities demonstrating the very high purity of the final products. The chimeric toxin migrated within somewhat lower than expected for 50 kDa protein M r range which may be related to the hydrophobicity of the molecule. The biologic activity of the rhIL-13 was exactly the same as commercially obtained hIL-13.
  • chimeric toxins such as hIL-13-PE38QQR
  • the cytotoxic activity of chimeric toxins were tested by measuring inhibition of protein synthesis. Protein synthesis was assayed by plating about 1 ⁇ 10 4 cells per in a 24-well tissue culture plate in 1 ml of medium. Various concentrations of the chimeric toxins were added 20-28 h following cell plating. After 20 h incubation with chimeric toxins, 3 H!-leucine was added to cells for 4 h, and the cell-associated radioactivity was measured. For blocking studies, rhIL-2, 4 or 13 was added to cells for 30 min before the chimeric toxin addition. Data were obtained from the average of duplicates and the assays were repeated several times.
  • hIL-13-PE38QQR cytotoxic to them.
  • cancers derived from colon, skin and stomach were examined.
  • the cancer cells were sensitive to hIL-13-PE38QQR with ID 50 s ranging from less than 1 ng/ml to 300 ng/ml (20 pM to 6.0 nM) (ID 50 indicates the concentration of the chimeric toxin at which the protein synthesis fell by 50% when compared to the sham-treated cells).
  • ID 50 indicates the concentration of the chimeric toxin at which the protein synthesis fell by 50% when compared to the sham-treated cells.
  • a colon adenocarcinoma cell line, Colo201 was very responsive with an IC 50 of 1 ng/ml.
  • A431 epidermoid carcinoma cells were also very sensitive to the action of hIL-13-toxin; the ID 50 for hIL-13-PE38QQR ranged from 6 to 10 ng/ml.
  • a gastric carcinoma CRL1739 cell line responded moderately to the hIL-13-toxin with an ID 50 of 50 ng/ml.
  • Another colon carcinoma cell line, Colo205 had a poorer response with an ID 50 of 300 ng/ml.
  • hIL-13-PE38QQR The cytotoxic action of hIL-13-PE38QQR was specific as it was blocked by a 10-fold excess of hIL-13 on all cells.
  • the hIL-13R has been suggested to share the ⁇ c subunit of the IL-2R (Russell et al. Science 262: 1880-1883 (1993))
  • the specificity of hIL-13-PE38QQR action on A431 and CRL1739 cells was further explored.
  • the cells were treated with hIL-13-PE38QQR with or without rhIL-2 at a concentration of 1.0 ⁇ g/ml or 10 ⁇ g/ml.
  • the rhIL-2 did not have any blocking action on hIL-13-PE38QQR on the two cell lines, even at 10,000 fold molar excess over the chimeric toxin.
  • IL-4 unlike IL-2, blocks the action of IL-13-PE38QOR
  • hIL-4 Native hIL-4 was added to cells which were then treated with hIL-13-PE38QQR. Unexpectedly, it was found that hIL-4 inhibited the cytotoxic activity of the hIL-13-toxin. This phenomenon was seen on all the tested cell lines, including Colo201, A431 and CRL1739. To investigate the possibility that hIL-13 and hIL-4 may compete for the same binding site, the cells were also treated with the hIL-4-based recombinant toxin, hIL-4-PE38QQR (Debinski et al. Int. J. Cancer 8: 744-748 (1994)).
  • hIL-4-PE38QQR The cytotoxic action of hIL-4-PE38QQR had already been shown to be blocked by an excess of hIL-4 but not of hIL-2 (Id.).
  • hIL-13 potently blocked the cytotoxic activity of hIL-4-PE38QQR.
  • hIL-4-PE4E another hIL-4-based chimeric toxin, hIL-4-PE4E (Debinski et al. J. Biol. Chem. 268: 14065-14070 (1993)), was blocked by an excess of hIL-13 on Colo201 and A431 cells.
  • hIL-13-PE38QQR cytotoxic action of hIL-13-PE38QQR is blocked by an excess of hIL-13 or hIL-4, and the cytotoxic action of hIL-4-PE38QQR is also blocked by the same two growth factors.
  • IL-2 does not block the action of either chimeric toxin.
  • binding ability of human IL-13 was compared to human IL-4-PE38QQR in competitive binding assays.
  • Recombinant hIL-4-PE38QQR was labeled with 125 I using the lactoperoxidase method as described by Debinski et al., J. Clin. Invest. 90, 405-411 (1992).
  • Binding assays were performed by a standard saturation and displacement curves analysis.
  • A431 epidermoid carcinoma cells were seeded at 10 5 cells per well in a 24-well tissue culture plates at 24 h before the experiment. The plates were placed on ice and cells were washed with ice-cold PBS without Ca++, Mg++ in 0.2% BSA, as described (Id.).
  • hIL-13 or hIL-4-PE38QQR Increasing concentrations of hIL-13 or hIL-4-PE38QQR were added to cells and incubated 30 min prior to the addition of fixed amount of 125 I-hIL-4-PE38QQR (specific activity 6.2 ⁇ Ci/ ⁇ g protein) for 2 to 3 h. After incubation, the cells were washed twice and lysed with 0.1N NaOH, and the radioactivity was counted in a ⁇ -counter.
  • Human IL-4-PE38QQR competed for the binding of 125 I-hIL-4-PE38QQR to A431 cells with an apparent ID 50 of 4 ⁇ 10 -8 M.
  • hIL-13 also competed for the 125 I-hIL-4-PE38QQR binding site with a comparable potency to that exhibited by the chimeric protein. More extensive binding studies have shown that hIL-13 also competes for hIL-4 binding sites on human renal carcinoma cell lines.
  • hIL-13 or hIL-4 The possibility of an influence of hIL-13 or hIL-4 on the process of receptor-mediated endocytosis and post-binding PE cellular toxicity steps was excluded by adding to cells: (i) native PE (PE binds to the ⁇ 2 -macroglobulin receptor), (ii) TGF ⁇ -PE40, and (iii) a recombinant immunotoxin C242rF(ab)-PE38QQR (Debinski et al. Clin. Res. 42, 251A, (Abstr.) (1994)). C242rF(ab)-PE38QQR binds a tumor-associated antigen that is a sialylated glycoprotein (Debinski et al. J. Clin. Invest. 90: 405-411 (1992)). The expected cytotoxic actions of these recombinant toxins were observed and neither hIL-13 nor hIL-4 blocked these actions on A431 and Colo205 cells
  • hIL-4 and hIL-13 compete for a common binding site on carcinoma cells but evoke different biological effects
  • hIL-13 and hIL-4 compete for a common binding site, they induce different cellular effects.
  • Protein synthesis was inhibited in A431 epidermoid carcinoma cells in a dose-dependent manner by hIL-4 alone, or by a ADP-ribosylation deficient chimeric toxin containing hIL-4 (Debinski et al. Int. J. Cancer 58: 744-748 (1994)).
  • This effect of hIL-4 or enzymatically deficient chimeric toxin can be best seen with a prolonged time of incubation ( ⁇ 24 h) and requires concentrations of hIL-4 many fold higher than that of the active chimeric toxin in order to cause a substantial decrease in tritium incorporation.
  • hIL-13 and hIL-4 may possess a common binding site, they appear to transduce differently in carcinoma cells expressing this common site, such as A431 and PM-RCC cells.

Abstract

This invention provides chimeric molecules useful for killing tumor cells bearing IL13 receptor(s) (IL-13R). The molecules comprise a cytotoxic molecule attached to a targeting molecule that specifically binds an IL-13 receptor. Preferred targeting molecules include IL-13 and anti-IL-13R antibodies.

Description

CROSS-REFERENCE TO RELATED APPLICATIONS
This is a Divisional of U.S. Ser. No. 08/404,685, filed on Mar. 15, 1995, now issued as U.S. Pat. No. 5,614,191.
FIELD OF THE INVENTION
This invention relates to methods of specifically delivering an effector molecule to a tumor cell. In particular this invention relates to chimeric molecules that specifically bind to IL-13 receptors and their use to deliver molecules having a particular activity to tumors overexpressing IL-13 receptors.
BACKGROUND OF THE INVENTION
In a chimeric molecule, two or more molecules that exist separately in their native state are joined together to form a single molecule having the desired functionality of all of its constituent molecules. Frequently, one of the constituent molecules of a chimeric molecule is a "targeting molecule". The targeting molecule is a molecule such as a ligand or an antibody that specifically binds to its corresponding target, for example a receptor on a cell surface. Thus, for example, where the targeting molecule is an antibody, the chimeric molecule will specifically bind (target) cells and tissues bearing the epitope to which the antibody is directed.
Another constituent of the chimeric molecule may be an "effector molecule". The effector molecule refers to a molecule that is to be specifically transported to the target to which the chimeric molecule is specifically directed. The effector molecule typically has a characteristic activity that is desired to be delivered to the target cell. Effector molecules include cytotoxins, labels, radionuclides, ligands, antibodies, drugs, liposomes, and the like.
In particular, where first effector molecule is a cytotoxin, the chimeric molecule may act as a potent cell-killing agent specifically targeting the cytotoxin to cells bearing a particular target molecule. For example, chimeric fusion proteins which include interleukin 4 (IL-4) or transforming growth factor (TGFα) fused to Pseudomonas exotoxin (PE) or interleukin 2 (IL-2) fused to Diphtheria toxin (DT) have been shown to specifically target and kill cancer cells (Pastan et al., Ann. Rev. Biochem., 61: 331-354 (1992)).
Generally, it is desirable to increase specificity and affinity and decrease cross-reactivity of the chimeric cytotoxins in order to increase their efficacy. To the extent the chimeric molecule preferentially selects and binds to its target (e.g. a tumor cell) and not to a non-target (e.g. a healthy cell), side effects of the chimeric molecule will be minimized. Unfortunately, many targets to which chimeric molecules have been directed (e.g. the IL-2 and IL-4 receptors), while showing elevated expression on tumor cells, are also expressed at significant levels on healthy cells. Thus, chimeric molecules directed to these targets (e.g. cytotoxins) show some adverse side-effects as they bind non-target cells that also express the targeted receptor.
SUMMARY OF THE INVENTION
The present invention provides methods and compositions for specifically delivering an effector molecule to a tumor cell. In particular, the present invention provides chimeric molecules that specifically target tumor cells with less binding to healthy cells than other analogous chimeric molecules known in the prior art.
The improved specific targeting of this invention is premised, in part, on the discovery that tumor cells, especially carcinomas such as renal cell carcinoma, overexpress IL-13 receptors at extremely high levels. The extremely high level of IL-13 receptor expression on target tumor cells permits the use of lower dosages of chimeric molecule to deliver the same amount of effector molecule to the target cells and also results in reduced binding of non-tumor cells.
In a preferred embodiment, this invention provides for a method for specifically delivering an effector molecule to a tumor cell bearing an IL-13 receptor. The method involves providing a chimeric molecule comprising an effector molecule attached to a targeting molecule that specifically binds to an IL-13 receptor and contacting the tumor with the chimeric molecule resulting in binding of the chimeric molecule to the tumor cell.
The targeting molecule is preferably either a ligand, such as IL-13, that specifically binds an IL-13 receptor or an anti-IL-13 receptor antibody. The targeting molecule may be conjugated to the effector molecule, or where both targeting and effector molecules are polypeptides, the targeting molecule may be joined to the effector molecule through one or more peptide bonds thereby forming a fusion protein. Suitable effector molecules include a cytotoxin, a label, a radionuclide, a drug, a liposome, a ligand, and an antibody. In a particularly preferred embodiment, the effector is a cytotoxin, more specifically a Pseudomonas exotoxin such as PE38QQR. Where the Pseudomonas exotoxin is fused to an IL-13 targeting molecule, a preferred fusion protein is IL-13-PE38QQR.
In another embodiment, this invention provides a method for impairing the growth of tumor cells, more preferably solid tumor cells, bearing an IL-13 receptor. The method involves contacting the tumor with a chimeric molecule comprising an effector molecule selected from the group consisting of a cytotoxin, a radionuclide, a ligand and an antibody; said effector molecule being attached to a targeting molecule that specifically binds a human IL-13 receptor. The targeting molecule is preferably a ligand (such as IL-13) that binds the IL-13 receptor or an anti-IL-13 receptor antibody. Preferred cytotoxic effector molecules include Pseudomonas exotoxin, Diphtheria toxin, ricin and abrin. Psuedomonas exotoxins, such as PE38QQR, are particularly preferred. The targeting molecule may be conjugated or fused to the effector molecule with attachment by fusion preferred for cytotoxic effector molecules. The tumor growth that is impaired may be tumor growth in a human. Thus the method may further comprise administering the chimeric molecule to a human intravenously into a body cavity, or into a human or an organ.
In yet another embodiment, this invention provides for a method of detecting the presence or absence of a tumor. The method involves contacting the tumor with a chimeric molecule comprising a detectable label attached to a targeting molecule that specifically binds a human IL-13 receptor and detecting the presence or absence of the label. In a preferred embodiment, the label is selected from the group consisting of a radioactive label, an enzymatic label, an electron dense label, and a fluorescent label.
This invention also provides for vectors comprising a nucleic acid sequence encoding a chimeric polypeptide fusion protein comprising an IL-13 attached to a second polypeptide. The chimeric polypeptide fusion protein specifically binds to a tumor cell bearing an IL-13 receptor. A preferred vector encodes an IL-13-PE fusion protein and more preferably encodes an IL-13-PE38QQR fusion protein.
This invention also provides for host cells comprising a nucleic acid sequence encoding a chimeric polypeptide fusion protein comprising an IL-13 attached to a second polypeptide. A preferred host cell comprises a nucleic acid encoding an IL-13-PE fusion protein, more preferably encoding an IL-13-PE38QQR fusion protein. The encoded fusion protein specifically binds to a tumor cell bearing an IL-13 receptor. Particularly preferred host cells are bacterial host cells, especially E. coli cells.
In still yet another embodiment, this invention provides chimeric molecules that specifically bind a tumor cell bearing an IL-13 receptor. In one preferred embodiment, the chimeric molecule comprises a cytotoxic molecule attached to a targeting molecule that specifically binds an IL-13. The targeting molecule may be conjugated or fused to the cytotoxic molecule. In a preferred embodiment, the targeting molecule is fused to the cytotoxin thereby forming a single-chain fusion protein. Particularly preferred targeting molecules are IL-13 or an antibody that specifically binds to the IL-13 receptor. Preferred cytotoxic molecules include Pseudomonas exotoxin, Diphtheria toxin, ricin, and abrin, with Pseudomonas exotoxins (especially PE38QQR) being most preferred.
In another preferred embodiment, the chimeric molecule comprises an effector molecule attached to an antibody that specifically binds to an IL-13 receptor. Effector molecules include a cytotoxin, a label, a radionuclide, a drug, liposome, a ligand and an antibody. The effector molecule may be fused or conjugated to the antibody.
The invention additionally provides for pharmacological compositions comprising a pharmaceutically acceptable carrier and a chimeric molecule where the chimeric molecule comprises and effector molecule attached to a targeting molecule that specifically binds to an IL-13 receptor. The targeting and effector molecules may be conjugated or fused to each other. Particularly preferred targeting molecules include IL-13 and anti-IL-13 receptor antibodies, while preferred effector molecules include a cytotoxin, a label, a radionuclide, a drug, a liposome, a ligand and an antibody. A preferred pharmacological composition includes an IL-13-PE fusion protein, more preferably an IL-13-PE38QQR fusion protein.
Definitions
The term "specifically deliver" as used herein refers to the preferential association of a molecule with a cell or tissue bearing a particular target molecule or marker and not to cells or tissues lacking that target molecule. It is, of course, recognized that a certain degree of non-specific interaction may occur between a molecule and a non-target cell or tissue. Nevertheless, specific delivery, may be distinguished as mediated through specific recognition of the target molecule. Typically specific delivery results in a much stronger association between the delivered molecule and cells bearing the target molecule than between the delivered molecule and cells lacking the target molecule. Specific delivery typically results in greater than 2 fold, preferably greater than 5 fold, more preferably greater than 10 fold and most preferably greater than 100 fold increase in amount of delivered molecule (per unit time) to a cell or tissue bearing the target molecule as compared to a cell or tissue lacking the target molecule or marker.
The term "residue" as used herein refers to an amino acid that is incorporated into a polypeptide. The amino acid may be a naturally occurring amino acid and, unless otherwise limited, may encompass known analogs of natural amino acids that can function in a similar manner as naturally occurring amino acids.
A "fusion protein" refers to a polypeptide formed by the joining of two or more polypeptides through a peptide bond formed between the amino terminus of one polypeptide and the carboxyl terminus of another polypeptide. The fusion protein may be formed by the chemical coupling of the constituent polypeptides or it may be expressed as a single polypeptide from nucleic acid sequence encoding the single contiguous fusion protein. A single chain fusion protein is a fusion protein having a single contiguous polypeptide backbone.
A "spacer" as used herein refers to a peptide that joins the proteins comprising a fusion protein. Generally a spacer has no specific biological activity other than to join the proteins or to preserve some minimum distance or other spatial relationship between them. However, the constituent amino acids of a spacer may be selected to influence some property of the molecule such as the folding, net charge, or hydrophobicity of the molecule.
A "ligand", as used herein, refers generally to all molecules capable of reacting with or otherwise recognizing or binding to a receptor on a target cell. Specifically, examples of ligands include, but are not limited to, antibodies, lymphokines, cytokines, receptor proteins such as CD4 and CD8, solubilized receptor proteins such as soluble CD4, hormones, growth factors, and the like which specifically bind desired target cells.
DETAILED DESCRIPTION
Chimeric Molecules Targeted to the IL-13 Receptor
The present invention provides a method for specifically delivering an effector molecule to a tumor cell. This method involves the use of chimeric molecules comprising a targeting molecule attached to an effector molecule. The chimeric molecules specifically target tumor cells while providing reduced binding to non-target cells as compared to other targeted chimeric molecules known in the art.
The improved specific targeting of this invention is premised, in part, on the discovery that solid tumors, especially carcinomas, overexpress IL-13 receptors at extremely high levels. While the IL-13 receptors are overexpressed on tumor cells, expression on other cells (e.g. monocytes and T cells) appears negligible. Thus, by specifically targeting the IL-13 receptor, the present invention provides chimeric molecules that are specifically directed to solid tumors while minimizing targeting of other cells or tissues.
In a preferred embodiment, this invention provides for compositions and methods for impairing the growth of tumors. The methods involve providing a chimeric molecule comprising a cytotoxic effector molecule attached to a targeting molecule that specifically binds an IL-13 receptor. The cytotoxin may be a native or modified cytotoxin such as Pseudomonas exotoxin (PE), Diphtheria toxin (DT), ricin, abrin, and the like.
The chimeric cytotoxin is administered to an organism containing tumor cells which are then contacted by the chimeric molecule. The targeting molecule component of the chimeric molecule specifically binds to the overexpressed IL-13 receptors on the tumor cells. Once bound to the IL-13 receptor on the cell surface, the cytotoxic effector molecule mediates internalization into the cell where the cytotoxin inhibits cellular growth or kills the cell.
The use of chimeric molecules comprising a targeting moiety joined to a cytotoxic effector molecules to target and kill tumor cells is known in the prior art. For example, chimeric fusion proteins which include interleukin 4 (IL-4) or transforming growth factor (TGFα) fused to Pseudomonas exotoxin (PE) or interleukin 2 (IL-2) fused to Diphtheria toxin (DT) have been tested for their ability to specifically target and kill cancer cells (Pastan et al., Ann. Rev. Biochem., 61: 331-354 (1992)).
Although chimeric IL-4-cytotoxin molecules are known in the prior art, and IL-4 shows some sequence similarity to IL-13, it was an unexpected discovery of the present invention that cytotoxins targeted by a moiety specific to the IL-13 receptor show significantly increased efficacy as compared to IL-4 receptor directed cytotoxins. Without being bound to a particular theory, it is believed that the improved efficacy of the IL-13 chimeras of the present invention is due to at least three factors.
First, IL-13 receptors are expressed at much lower levels, if at all on non-tumor cells (e.g. monocytes, T cells, B cells). Thus cytotoxins directed to IL-13 receptors show reduced binding and subsequent killing of healthy cells and tissues as compared to cytotoxins directed to IL-4 receptors.
Second, the receptor component that specifically binds IL-13 appears to be expressed at significantly higher levels on solid tumors than the receptor component that binds IL-4. Thus, tumor cells bind higher levels of cytotoxic chimeric molecules directed against IL-13 receptors than cytotoxic chimeric molecules directed against IL-4 receptors.
Finally, IL-4 receptors are up-regulated when immune system cells (e.g. T-cells) are activated. This results in healthy cells, for example T-cells and B-cells, showing greater susceptibility to IL-4 receptor directed cytotoxins. Thus, the induction of an immune reaponse (as against a cancer), results in greater susceptiblity of cells of the immune system to the therapeutic agent. In contrast, IL-13 receptors are not up-regulated in activated cells. Thus IL-13 receptor targeted cytotoxins have no greater effect on activated cells and thereby minimize adverse effects of the therapeutic composition on cells of the immune system.
In another embodiment, this invention also provides for compositions and methods for detecting the presence or absence of tumor cells. These methods involve providing a chimeric molecule comprising an effector molecule, that is a detectable label attached to a targeting molecule that specifically binds an IL-13 receptor. The IL-13 receptor targeting moiety specifically binds the chimeric molecule to tumor cells which are then marked by their association with the detectable label. Subsequent detection of the cell-associated label indicates the presence of a tumor cell.
In yet another embodiment, the effector molecule may be another specific binding moiety such as an antibody, a growth factor, or a ligand. The chimeric molecule will then act as a highly specific bifunctional linker. This linker may act to bind and enhance the interaction between cells or cellular components to which the fusion protein binds. Thus, for example, where the "targeting" component of the chimeric molecule comprises a polypeptide that specifically binds to an IL-13 receptor and the "effector" component is an antibody or antibody fragment (e.g. an Fv fragment of an antibody), the targeting component specifically binds cancer cells, while the effector component binds receptors (e.g., IL-2 or IL-4 receptors) on the surface of immune cells. The chimeric molecule may thus act to enhance and direct an immune response toward target cancer cells.
In still yet another embodiment the effector molecule may be a pharmacological agent (e.g. a drug) or a vehicle containing a pharmacological agent. This is particularly suitable where it is merely desired to invoke a non-lethal biological response. Thus the moiety that specifically binds to an IL-13 receptor may be conjugated to a drug such as vinblastine, doxirubicin, genistein (a tyrosine kinase inhibitor), an antisense molecule, and other pharmacological agents known to those of skill in the art, thereby specifically targeting the pharmacological agent to tumor cells over expressing IL-13 receptors.
Alternatively, the targeting molecule may be bound to a vehicle containing the therapeutic composition. Such vehicles include, but are not limited to liposomes, micelles, various synthetic beads, and the like.
One of skill in the art will appreciate that the chimeric molecules of the present invention may include multiple targeting moieties bound to a single effector or conversely, multiple effector molecules bound to a single targeting moiety. In still other embodiment, the chimeric molecules may include both multiple targeting moieties and multiple effector molecules. Thus, for example, this invention provides for "dual targeted" cytotoxic chimeric molecules in which targeting molecule that specifically binds to IL-13 is attached to a cytotoxic molecule and another molecule (e.g. an antibody, or another ligand) is attached to the other terminus of the toxin. Such a dual-targeted cytotoxin might comprise an IL-13 substituted for domain Ia at the amino terminus of a PE and anti-TAC(Fv) inserted in domain III, between amino acid 604 and 609. Other antibodies may also be suitable.
The Targeting Molecule
In a preferred embodiment, the targeting molecule is a molecule that specifically binds to the IL-13 receptor. The term "specifically binds", as used herein, when referring to a protein or polypeptide, refers to a binding reaction which is determinative of the presence of the protein or polypeptide in a heterogeneous population of proteins and other biologics. Thus, under designated conditions (e.g. immunoassay conditions in the case of an antibody), the specified ligand or antibody binds to its particular "target" protein (e.g. an IL-13 receptor protein) and does not bind in a significant amount to other proteins present in the sample or to other proteins to which the ligand or antibody may come in contact in an organism.
A variety of immunoassay formats may be used to select antibodies specifically immunoreactive with an IL-13 receptor protein. For example, solid-phase ELISA immunoassays are routinely used to select monoclonal antibodies specifically immunoreactive with a protein. See Harlow and Lane (1988) Antibodies, A Laboratory Manual, Cold Spring Harbor Publications, New York, for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity.
Similarly, assay formats for detecting specific binding of ligands (e.g. IL-13) with their respective receptor are also well known in the art. Example 1 provides a detailed protocol for assessing specific binding of labeled IL-13 by and IL-13 receptor.
The IL-13 receptor is a cell surface receptor that specifically binds IL-13 and mediates a variety of physiological responses in various cell types as described below in the description of IL-13. The IL-13 receptor may be identified by contacting a cell or other sample with labeled IL-13 and detecting the amount of specific binding of IL-13 according to methods well known to those of skill in the art. Detection of IL-13 receptors by labeled IL-13 binding is described in detail in Example 1.
Alternatively, an anti-IL-13 receptor antibody may also be used to identify IL-13 receptors. The antibody will specifically bind to the IL-13 receptor and this binding may be detected either through detection of a conjugated label or through detection of a labeled second antibody that binds the anti-IL-13 receptor antibody.
In a preferred embodiment, the moiety utilized to specifically target the IL-13 receptor is either an antibody that specifically binds the IL-13 receptor (an anti-IL-13R antibody) or a ligand, such as IL-13, that specifically binds to the receptor.
IL-13
IL-13 is a pleiotropic cytokine that is recognized to share many of the properties of IL-4. IL-13 has approximately 30% sequence identity with IL-4 and exhibits IL-4-like activities on monocytes/macrophages and human B cells (Minty et al., Nature, 362: 248 (1993), McKenzie et al. Proc. Natl. Acad. Sci. USA, 90: 3735 (1987)). In particular, IL-13 appears to be a potent regulator of inflammatory and immune responses. Like IL-4, IL-13 can up-regulate the monocyte/macrophage expression of CD23 and MHC class I and class II antigens, down-regulate the expression of Fcγ, and inhibit antibody-dependent cytotoxicity. IL-13 can also inhibit nitric oxide production as well as the expression of pro-inflammatory cytokines (e.g. IL-1, IL-6, IL-8, IL-10 and IL-12) and chemokines (MIP-1, MCP), but enhance the production of IL-1ra (Minty supra.; Mckenzie et al., supra.; Zurawski et al. Immunol. Today, 15: 19 (1994); de Wall Malefyt et al. J. Immunol., 150: 180A (1993); de Wall Malefyt et al. J. Immunol., 151: 6370 (1993); Doherty et al. J. Immunol., 151: 7151 (1993); and Minty et al. Eur. cytokine Netw., 4: 99 (1993)).
Recombinant IL-13 is commercially available from a number of sources (see, e.g. R & D Systems, Minneapolis, Minn., USA, and Sanofi Bio-Industries, Inc., Tervose, Pa., USA). Alternatively, a gene or a cDNA encoding IL-13 may be cloned into a plasmid or other expression vector and expressed in any of a number of expression systems according to methods well known to those of skill in the art. Methods of cloning and expressing IL-13 and the nucleic acid sequence for IL-13 are well kown (see, for example, Minty et al. (1993) supra. and McKenzie (1987), supra). In addition, the expression of IL-13 as a component of a chimeric molecule is detailed in Example 4.
One of skill in the art will appreciate that analogues or fragments of IL-13 bearing will also specifically bind to the IL-13 receptor. For example, conservative substitutions of residues (e.g., a serine for an alanine or an aspartic acid for a glutamic acid) comprising native IL-13 will provide IL-13 analogues that also specifically bind to the IL-13 receptor. Thus, the term "IL-13", when used in reference to a targeting molecule, also includes fragments, analogues or peptide mimetics of IL-13 that also specifically bind to the IL-13 receptor.
Anti-IL-13 Receptor Antibodies
One of skill will recognize that other molecules besides IL-13 will specifically bind to IL-13 receptors. Polyclonal and monoclonal antibodies directed against IL-13 receptors provide particularly suitable targeting molecules in the chimeric molecules of this invention. The term "antibody", as used herein, includes various forms of modified or altered antibodies, such as an intact immunoglobulin, various fragments such as an Fv fragment, an Fv fragment containing only the light and heavy chain variable regions, an Fv fragment linked by a disulfide bond (Brinkmann, et al. Proc. Natl. Acad. Sci. USA, 90: 547-551 (1993)), an Fab or (Fab)'2 fragment containing the variable regions and parts of the constant regions, a single-chain antibody and the like (Bird et al., Science 242: 424-426 (1988); Huston et al., Proc. Nat. Acad. Sci. USA 85: 5879-5883 (1988)). The antibody may be of animal (especially mouse or rat) or human origin or may be chimeric (Morrison et al., Proc Nat. Acad. Sci. USA 81: 6851-6855 (1984)) or humanized (Jones et al., Nature 321: 522-525 (1986), and published UK patent application #8707252). Methods of producing antibodies suitable for use in the present invention are well known to those skilled in the art and can be found described in such publications as Harlow & Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory (1988), and Asai, Methods in Cell Biology Vol. 37: Antibodies in Cell Biology, Academic Press, Inc. N.Y. (1993).
Antibodies that specifically bind the IL-13 receptor may be produced by a number of means well known to those of skill in the art. Generally, this involves using an antigenic component of the IL-13 receptor as an antigen to induce the production of antibodies in an organism (e.g. a sheep, mouse, rabbit, etc.). One of skill in the art will recognize that there are numerous methods of isolating all or components of the IL-13 receptor for use as an antigen. For example, IL-13 receptors may be isolated by cross-linking the receptor to a labeled IL-13 by the exposure to 2 mM disuccinimidyl suberate (DSS). The labeled receptor may then be isolated according to routine methods and the isolated receptor may be used as an antigen to raise anti-IL-13 receptor antibodies as described below. Cross-linking and isolation of components of the IL-13 receptor is described in Example 3.
In a preferred embodiment, however, IL-13 receptors may be isolated by means of affinity chromatography. It was a surprising discovery of the present invention that solid tumor cells overexpress IL-13 receptors. This discovery of cells overexpressing IL-13 receptor greatly simplifies the receptor isolation. Generally, approximately, 100 million renal carcinoma cells, may be solubilized in detergent with protease inhibitors according to standard methods. The resulting lysate is then run through an affinity column bearing IL-13. The receptor binds to the IL-13 in the column thereby effecting an isolation from the lysate. The column is then eluted with a low pH buffer to dissociate the IL-13 ligand from the IL-13 receptor resulting in isolated receptor. The isolated receptor may then be used as an antigen to raise anti-IL-13 receptor antibodies.
Antibody Production
Methods of producing polyclonal antibodies are known to those of skill in the art. In brief, an immunogen, preferably an isolated IL-13 receptor or receptor epitope is mixed with an adjuvant and animals are immunized with the mixture. The animal's immune response to the immunogen preparation is monitored by taking test bleeds and determining the titer of reactivity to the polypeptide of interest. When appropriately high titers of antibody to the immunogen are obtained, blood is collected from the animal and antisera are prepared. Further fractionation of the antisera to enrich for antibodies reactive to the polypeptide is performed where desired. See, e.g., Coligan (1991) Current Protocols in Immunology Wiley/Greene, New York; and Harlow and Lane (1989) Antibodies: A Laboratory Manual Cold Spring Harbor Press, New York, which are incorporated herein by reference.
Monoclonal antibodies may be obtained by various techniques familiar to those skilled in the art. Description of techniques for preparing such monoclonal antibodies may be found in, e.g., Stites et al. (eds.) Basic and Clinical Immunology (4th ed.) Lange Medical Publications, Los Altos, Calif., and references cited therein; Harlow and Lane (1988) Antibodies: A Laboratory Manual CSH Press; Goding (1986) Monoclonal Antibodies: Principles and Practice (2d ed.) Academic Press, New York, N.Y.; and particularly in Kohler and Milstein (1975) Nature 256: 495-497, which discusses one method of generating monoclonal antibodies.
Summarized briefly, this method involves injecting an animal with an immunogen. The animal is then sacrificed and cells taken from its spleen, which are then fused with myeloma cells (See, Kohler and Milstein (1976) Eur. J. Immunol. 6: 511-519, incorporated herein by reference). The result is a hybrid cell or "hybridoma" that is capable of reproducing in vitro.
Colonies arising from single immortalized cells are screened for production of antibodies of the desired specificity and affinity for the antigen, and yield of the monoclonal antibodies produced by such cells is enhanced by various techniques, including injection into the peritoneal cavity of a vertebrate host. Alternatively, one may isolate DNA sequences which encode a monoclonal antibody or a binding fragment thereof by screening a DNA library from human B cells according to the general protocol outlined by Huse et al. (1989) Science 246: 1275-1281. In this manner, the individual antibody species obtained are the products of immortalized and cloned single B cells from the immune animal generated in response to a specific site recognized on the immunogenic substance.
Other suitable techniques involve selection of libraries of antibodies in phage or similar vectors. See, Huse et al. Science 246: 1275-1281 (1989); and Ward, et al. Nature 341: 544-546 (1989). In general suitable monoclonal antibodies will usually bind their target epitope with at least a KD of about 1 mM, more usually at least about 300 μM, and most preferably at least about 0.1 μM or better.
Other Targeting Antibodies
Where the chimeric molecule contains more than one targeting molecule (e.g. a dual-targeted cytotoxin), the molecule may contain targeting antibodies directed to tumor markers other than the overexpressed IL-13 receptor. A number of such antibodies are known and have even been converted to form suitable for incorporation into fusion proteins. These include anti-erbB2, B3, BR96, OVB3, anti-transferrin, Mik-β1 and PR1 (see Batra et al., Mol. Cell. Biol., 11: 2200-2205 (1991); Batra et al., Proc. Natl. Acad. Sci. USA, 89: 5867-5871 (1992); Brinkmann, et al. Proc. Natl. Acad. Sci. USA, 88: 8616-8620 (1991); Brinkmann et al., Proc. Natl. Acad. Sci. USA, 90: 547-551 (1993); Chaudhary et al., Proc. Natl. Acad. Sci. USA, 87: 1066-1070 (1990); Friedman et al., Cancer Res. 53: 334-339 (1993); Kreitman et al., J. Immunol., 149: 2810-2815 (1992); Nicholls et al., J. Biol. Chem., 268: 5302-5308 (1993); and Wells, et al., Cancer Res., 52: 6310-6317 (1992), respectively).
The Effector Molecule
As described above, the effector molecule component of the chimeric molecules of this invention may be any molecule whose activity it is desired to deliver to cells that overexpress IL-13 receptors. Particularly preferred effector molecules include cytotoxins such as PE or DT, radionuclides, ligands such as growth factors, antibodies, detectable labels such as fluorescent or radioactive labels, and therapeutic compositions such as liposomes and various drugs.
Cytotoxins
Particularly preferred cytotoxins include Pseudomonas exotoxins, Diphtheria toxins, ricin, and abrin. Pseudomonas exotoxin and Dipthteria toxin are most preferred.
Pseudomonas exotoxin (PE)
Pseudomonas exotoxin A (PE) is an extremely active monomeric protein (molecular weight 66 kD), secreted by Pseudomonas aeruginosa, which inhibits protein synthesis in eukaryotic cells through the inactivation of elongation factor 2 (EF-2) by catalyzing its ADP-ribosylation (catalyzing the transfer of the ADP ribosyl moiety of oxidized NAD onto EF-2).
The toxin contains three structural domains that act in concert to cause cytotoxicity. Domain Ia (amino acids 1-252) mediates cell binding. Domain II (amino acids 253-364) is responsible for translocation into the cytosol and domain III (amino acids 400-613) mediates ADP ribosylation of elongation factor 2, which inactivates the protein and causes cell death. The function of domain Ib (amino acids 365-399) remains undefined, although a large part of it, amino acids 365-380, can be deleted without loss of cytotoxicity. See Siegall et al., J. Biol. Chem. 264: 14256-14261 (1989), incorporated by reference herein.
Where the targeting molecule (e.g. IL-13) is fused to PE, a preferred PE molecule is one in which domain Ia (amino acids 1 through 252) is deleted and amino acids 365 to 380 have been deleted from domain Ib. However all of domain Ib and a portion of domain II (amino acids 350 to 394) can be deleted, particularly if the deleted sequences are replaced with a linking peptide such as GGGGS (SEQ ID NO:1).
In addition, the PE molecules can be further modified using site-directed mutagenesis or other techniques known in the art, to alter the molecule for a particular desired application. Means to alter the PE molecule in a manner that does not substantially affect the functional advantages provided by the PE molecules described here can also be used and such resulting molecules are intended to be covered herein.
For maximum cytotoxic properties of a preferred PE molecule, several modifications to the molecule are recommended. An appropriate carboxyl terminal sequence to the recombinant molecule is preferred to translocate the molecule into the cytosol of target cells. Amino acid sequences which have been found to be effective include, REDLK (SEQ ID NO:2) (as in native PE), REDL (SEQ ID NO:3), RDEL (SEQ ID NO:4), or KDEL (SEQ ID NO:5), repeats of those, or other sequences that function to maintain or recycle proteins into the endoplasmic reticulum, referred to here as "endoplasmic retention sequences". See, for example, Chaudhary et al, Proc. Natl. Acad. Sci. USA 87:308-312 and Seetharam et al, J. Biol. Chem. 266: 17376-17381 (1991) and commonly assigned, U.S. Ser. No. 07/459,635 filed Jan. 2, 1990, all of which are incorporated by reference herein.
Deletions of amino acids 365-380 of domain Ib can be made without loss of activity. Further, a substitution of methionine at amino acid position 280 in place of glycine to allow the synthesis of the protein to begin and of serine at amino acid position 287 in place of cysteine to prevent formation of improper disulfide bonds is beneficial. In a preferred embodiment, the targeting molecule is inserted in replacement for domain Ia. A similar insertion has been accomplished in what is known as the TGFα-PE40 molecule (also referred to as TP40) described in Heimbrook et al., Proc. Natl. Acad. Sci., USA, 87: 4697-4701 (1990) and in commonly assigned U.S. Ser. No. 07/865,722 filed Apr. 8, 1992 and in U.S. Ser. No. 07/522,563 filed May 14, 1990, all of which are incorporated by reference.
Preferred forms of PE contain amino acids 253-364 and 381-608, and are followed by the native sequences REDLK (SEQ ID NO:2) or the mutant sequences KDEL (SEQ ID NO:5) or RDEL (SEQ ID NO:4).
Lysines at positions 590 and 606 may or may not be mutated to glutamine.
In a particularly preferred embodiment, the IL-13 receptor targeted cytotoxins of this invention comprise the PE molecule designated PE38QQR. This PE molecule is a truncated form of PE composed of amino acids 253-364 and 381-608. The lysine residues at positions 509 and 606 are replaced by glutamine and at 613 are replaced by arginine (Debinski et al. Bioconj. Chem., 5: 40 (1994) which is incorporated herein by reference).
The targeting molecule (e.g. IL-13 or anti-IL-13R antibody) may also be inserted at a point within domain III of the PE molecule. Most preferably the targeting molecule is fused between about amino acid positions 607 and 609 of the PE molecule. This means that the targeting molecule is inserted after about amino acid 607 of the molecule and an appropriate carboxyl end of PE is recreated by placing amino acids about 604-613 of PE after the targeting molecule. Thus, the targeting molecule is inserted within the recombinant PE molecule after about amino acid 607 and is followed by amino acids 604-613 of domain III. The targeting molecule may also be inserted into domain Ib to replace sequences not necessary for toxicity. Debinski, et al. Mol. Cell. Biol., 11: 1751-1753 (1991).
In a preferred embodiment, the PE molecules will be fused to the targeting molecule by recombinant means. The genes encoding protein chains may be cloned in cDNA or in genomic form by any cloning procedure known to those skilled in the art. See for example Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, (1989), incorporated by reference herein. Methods of cloning genes encoding PE fused to various ligands are well known to those of skill in the art. See, for example, Siegall et al., FASEB J., 3: 2647-2652 (1989); Chaudhary et al. Proc. Natl. Acad. Sci. USA, 84: 4538-4542 (1987), which are incorporated herein by reference.
Those skilled in the art will realize that additional modifications, deletions, insertions and the like may be made to the chimeric molecules of the present invention or to the nucleic acid sequences encoding IL-13 receptor-directed chimeric molecules. Especially, deletions or changes may be made in PE or in a linker connecting an antibody gene to PE, in order to increase cytotoxicity of the fusion protein toward target cells or to decrease nonspecific cytotoxicity toward cells without antigen for the antibody. All such constructions may be made by methods of genetic engineering well known to those skilled in the art (see, generally, Sambrook et al., supra) and may produce proteins that have differing properties of affinity, specificity, stability and toxicity that make them particularly suitable for various clinical or biological applications.
Diphtheria Toxin (DT)
Like PE, diphtheria toxin (DT) kills cells by ADP-ribosylating elongation factor 2 thereby inhibiting protein synthesis. Diphtheria toxin, however, is divided into two chains, A and B, linked by a disulfide bridge. In contrast to PE, chain B of DT, which is on the carboxyl end, is responsible for receptor binding and chain A, which is present on the amino end, contains the enzymatic activity (Uchida et al., Science, 175: 901-903 (1972); Uchida et al. J. Biol. Chem., 248: 3838-3844 (1973)).
In a preferred embodiment, the targeting molecule-Diphtheria toxin fusion proteins of this invention have the native receptor-binding domain removed by truncation of the Diphtheria toxin B chain. Particularly preferred is DT388, a DT in which the carboxyl terminal sequence beginning at residue 389 is removed. Chaudhary, et al., Bioch. Biophys. Res. Comm., 180: 545-551 (1991).
Like the PE chimeric cytotoxins, the DT molecules may be chemically conjugated to the IL-13 receptor targeting molecule, but, in a preferred embodiment, the targeting molecule will be fused to the Diphtheria toxin by recombinant means. The genes encoding protein chains may be cloned in cDNA or in genomic form by any cloning procedure known to those skilled in the art. Methods of cloning genes encoding DT fused to various ligands are also well known to those of skill in the art. See, for example, Williams et al. J. Biol. Chem. 265: 11885-11889 (1990) and copending patent application (U.S. Ser. No. 07/620,939) which describe the expression of a number of growth-factor-DT fusion proteins.
The term "Diphtheria toxin" (DT) as used herein refers to full length native DT or to a DT that has been modified. Modifications typically include removal of the targeting domain in the B chain and, more specifically, involve truncations of the carboxyl region of the B chain.
Detectable Labels
Detectable labels suitable for use as the effector molecule component of the chimeric molecules of this invention include any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means. Useful labels in the present invention include magnetic beads (e.g. DYNABEADS™), fluorescent dyes (e.g., fluorescein isothiocyanate, texas red, rhodamine, green fluorescent protein, and the like), radiolabels (e.g., 3 H, 125 I, 35 S, 14 C, or 32P), enzymes (e.g., horse radish peroxidase, alkaline phosphatase and others commonly used in an ELISA), and colorimetric labels such as colloidal gold or colored glass or plastic (e.g. polystyrene, polypropylene, latex, etc.) beads.
Means of detecting such labels are well known to those of skill in the art. Thus, for example, radiolabels may be detected using photographic film or scintillation counters, fluorescent markers may be detected using a photodetector to detect emitted illumination. Enzymatic labels are typically detected by providing the enzyme with a substrate and detecting the reaction product produced by the action of the enzyme on the substrate, and colorimetric labels are detected by simply visualizing the colored label.
Ligands
As explained above, the effector molecule may also be a ligand or an antibody. Particularly preferred ligand and antibodies are those that bind to surface markers on immune cells. Chimeric molecules utilizing such antibodies as effector molecules act as bifunctional linkers establishing an association between the immune cells bearing binding partner for the ligand or antibody and the tumor cells overexpressing the IL-13 receptor. Suitable antibodies and growth factors are known to those of skill in the art and include, but are not limited to, IL-2, IL-4, IL-6, IL-7, tumor necrosis factor (TNF), anti-Tac, TGFα, and the like.
Other Therapeutic Moieties
Other suitable effector molecules include pharmacological agents or encapsulation systems containing various pharmacological agents. Thus, the targeting molecule of the chimeric molecule may be attached directly to a drug that is to be delivered directly to the tumor. Such drugs are well known to those of skill in the art and include, but are not limited to, doxirubicin, vinblastine, genistein, an antisense molecule, and the like.
Alternatively, the effector molecule may be an encapsulation system, such as a liposome or micelle that contains a therapeutic composition such as a drug, a nucleic acid (e.g. an antisense nucleic acid), or another therapeutic moiety that is preferably shielded from direct exposure to the circulatory system. Means of preparing liposomes attached to antibodies are well known to those of skill in the art. See, for example, U.S. Pat. No. 4,957,735, Connor et al., Pharm. Ther., 28: 341-365 (1985).
Attachment of the Targeting Molecule to the Effector Molecule
One of skill will appreciate that the targeting molecule and effector molecules may be joined together in any order. Thus, where the targeting molecule is a polypeptide, the effector molecule may be joined to either the amino or carboxy termini of the targeting molecule. The targeting molecule may also be joined to an internal region of the effector molecule, or conversely, the effector molecule may be joined to an internal location of the targeting molecule, as long as the attachment does not interfere with the respective activities of the molecules.
The targeting molecule and the effector molecule may be attached by any of a number of means well known to those of skill in the art. Typically the effector molecule is conjugated, either directly or through a linker (spacer), to the targeting molecule. However, where both the effector molecule and the targeting molecule are polypeptides it is preferable to recombinantly express the chimeric molecule as a single-chain fusion protein.
Conjugation of the Effector Molecule to the Targeting Molecule
In one embodiment, the targeting molecule (e.g. IL-13 or anti-IL-13R antibody) is chemically conjugated to the effector molecule (e.g. a cytotoxin, a label, a ligand, or a drug or liposome). Means of chemically conjugating molecules are well known to those of skill.
The procedure for attaching an agent to an antibody or other polypeptide targeting molecule will vary according to the chemical structure of the agent. Polypeptides typically contain variety of functional groups; e.g., carboxylic acid (COOH) or free amine (--NH2) groups, which are available for reaction with a suitable functional group on an effector molecule to bind the effector thereto.
Alternatively, the targeting molecule and/or effector molecule may be derivatized to expose or attach additional reactive functional groups. The derivatization may involve attachment of any of a number of linker molecules such as those available from Pierce Chemical Company, Rockford Ill.
A "linker", as used herein, is a molecule that is used to join the targeting molecule to the effector molecule. The linker is capable of forming covalent bonds to both the targeting molecule and to the effector molecule. Suitable linkers are well known to those of skill in the art and include, but are not limited to, straight or branched-chain carbon linkers, heterocyclic carbon linkers, or peptide linkers. Where the targeting molecule and the effector molecule are polypeptides, the linkers may be joined to the constituent amino acids through their side groups (e.g., through a disulfide linkage to cysteine). However, in a preferred embodiment, the linkers will be joined to the alpha carbon amino and carboxyl groups of the terminal amino acids.
A bifunctional linker having one functional group reactive with a group on a particular agent, and another group reactive with an antibody, may be used to form the desired immunoconjugate. Alternatively, derivatization may involve chemical treatment of the targeting molecule, e.g., glycol cleavage of the sugar moiety of a the glycoprotein antibody with periodate to generate free aldehyde groups. The free aldehyde groups on the antibody may be reacted with free amine or hydrazine groups on an agent to bind the agent thereto. (See U.S. Pat. No. 4,671,958). Procedures for generation of free sulfhydryl groups on polypeptide, such as antibodies or antibody fragments, are also known (See U.S. Pat. No. 4,659,839).
Many procedure and linker molecules for attachment of various compounds including radionuclide metal chelates, toxins and drugs to proteins such as antibodies are known. See, for example, European Patent Application No. 188,256; U.S. Pat. Nos. 4,671,958, 4,659,839, 4,414,148, 4,699,784; 4,680,338; 4,569,789; and 4,589,071; and Borlinghaus et al. Cancer Res. 47: 4071-4075 (1987) which are incorporated herein by reference. In particular, production of various immunotoxins is well-known within the art and can be found, for example in "Monoclonal Antibody-Toxin Conjugates: Aiming the Magic Bullet," Thorpe et al., Monoclonal Antibodies in Clinical Medicine, Academic Press, pp. 168-190 (1982), Waldmann, Science, 252: 1657 (1991), U.S. Pat. Nos. 4,545,985 and 4,894,443 which are incorporated herein by reference.
In some circumstances, it is desirable to free the effector molecule from the targeting molecule when the chimeric molecule has reached its target site. Therefore, chimeric conjugates comprising linkages which are cleavable in the vicinity of the target site may be used when the effector is to be released at the target site. Cleaving of the linkage to release the agent from the antibody may be prompted by enzymatic activity or conditions to which the immunoconjugate is subjected either inside the target cell or in the vicinity of the target site. When the target site is a tumor, a linker which is cleavable under conditions present at the tumor site (e.g. when exposed to tumor-associated enzymes or acidic pH) may be used.
A number of different cleavable linkers are known to those of skill in the art. See U.S. Pat. Nos. 4,618,492; 4,542,225, and 4,625,014. The mechanisms for release of an agent from these linker groups include, for example, irradiation of a photolabile bond and acid-catalyzed hydrolysis. U.S. Pat. No. 4,671,958, for example, includes a description of immunoconjugates comprising linkers which are cleaved at the target site in vivo by the proteolytic enzymes of the patient's complement system. In view of the large number of methods that have been reported for attaching a variety of radiodiagnostic compounds, radiotherapeutic compounds, drugs, toxins, and other agents to antibodies one skilled in the art will be able to determine a suitable method for attaching a given agent to an antibody or other polypeptide.
Production of Fusion Proteins
Where the targeting molecule and/or the effector molecule is relatively short (i.e., less than about 50 amino acids) they may be synthesized using standard chemical peptide synthesis techniques. Where both molecules are relatively short the chimeric molecule may be synthesized as a single contiguous polypeptide. Alternatively the targeting molecule and the effector molecule may be synthesized separately and then fused by condensation of the amino terminus of one molecule with the carboxyl terminus of the other molecule thereby forming a peptide bond. Alternatively, the targeting and effector molecules may each be condensed with one end of a peptide spacer molecule thereby forming a contiguous fusion protein.
Solid phase synthesis in which the C-terminal amino acid of the sequence is attached to an insoluble support followed by sequential addition of the remaining amino acids in the sequence is the preferred method for the chemical synthesis of the polypeptides of this invention. Techniques for solid phase synthesis are described by Barany and Merrifield, Solid-Phase Peptide Synthesis; pp. 3-284 in The Peptides: Analysis, Synthesis, Biology. Vol. 2: Special Methods in Peptide Synthesis, Part A., Merrifield, et al. J. Am. Chem. Soc., 85: 2149-2156 (1963), and Stewart et al., Solid Phase Peptide Synthesis, 2nd ed. Pierce Chem. Co., Rockford, Ill. (1984) which are incorporated herein by reference.
In a preferred embodiment, the chimeric fusion proteins of the present invention are synthesized using recombinant DNA methodology. Generally this involves creating a DNA sequence that encodes the fusion protein, placing the DNA in an expression cassette under the control of a particular promoter, expressing the protein in a host, isolating the expressed protein and, if required, renaturing the protein.
DNA encoding the fusion proteins (e.g. IL-13-PE38QQR) of this invention may be prepared by any suitable method, including, for example, cloning and restriction of appropriate sequences or direct chemical synthesis by methods such as the phosphotriester method of Narang et al. Meth. Enzymol. 68: 90-99 (1979); the phosphodiester method of Brown et al., Meth. Enzymol. 68: 109-151 (1979); the diethylphosphoramidite method of Beaucage et al., Tetra. Lett., 22: 1859-1862 (1981); and the solid support method of U.S. Pat. No. 4,458,066, all incorporated by reference herein.
Chemical synthesis produces a single stranded oligonucleotide. This may be converted into double stranded DNA by hybridization with a complementary sequence, or by polymerization with a DNA polymerase using the single strand as a template. One of skill would recognize that while chemical synthesis of DNA is limited to sequences of about 100 bases, longer sequences may be obtained by the ligation of shorter sequences.
Alternatively, subsequences may be cloned and the appropriate subsequences cleaved using appropriate restriction enzymes. The fragments may then be ligated to produce the desired DNA sequence.
In a preferred embodiment, DNA encoding fusion proteins of the present invention may be cloned using DNA amplification methods such as polymerase chain reaction (PCR). Thus, in a preferred embodiment, the gene for IL-13 is PCR amplified, using a sense primer containing the restriction site for NdeI and an antisense primer containing the restriction site for HindIII. In a particularly preferred embodiment, the primers are selected to amplify the nucleic acid starting at position 19, as described by McKenzie et al. (1987), supra. This produces a nucleic acid encoding the mature IL-13 sequence and having terminal restriction sites. A PE38QQR fragment may be cut out of the plasmid pWDMH4-38QQR or plasmid pSGC242FdN1 described by Debinski et al. Int. J. Cancer, 58: 744-748 (1994), and by Debinski et al. Clin. Res., 42: 251A (abstract (1994) respectively. Ligation of the IL-13 and PE38QQR sequences and insertion into a vector produces a vector encoding IL-13 joined to the amino terminus of PE38QQR (position 253 of PE). The two molecues are joined by a three amino acid junction consaisting of glutamic acid, alanine, and phenylalanine introduced by the restriction site.
While the two molecules are preferrably essentially directly joined together, one of skill will appreciate that the molecules may be separated by a peptide spacer consisting of one or more amino acids. Generally the spacer will have no specific biological activity other than to join the proteins or to preserve some minimum distance or other spatial relationship between them. However, the constituent amino acids of the spacer may be selected to influence some property of the molecule such as the folding, net charge, or hydrophobicity.
The nucleic acid sequences encoding the fusion proteins may be expressed in a variety of host cells, including E. coli, other bacterial hosts, yeast, and various higher eukaryotic cells such as the COS, CHO and HeLa cells lines and myeloma cell lines. The recombinant protein gene will be operably linked to appropriate expression control sequences for each host. For E. coli this includes a promoter such as the T7, trp, or lambda promoters, a ribosome binding site and preferably a transcription termination signal. For eukaryotic cells, the control sequences will include a promoter and preferably an enhancer derived from immunoglobulin genes, SV40, cytomegalovirus, etc., and a polyadenylation sequence, and may include splice donor and acceptor sequences.
The plasmids of the invention can be transferred into the chosen host cell by well-known methods such as calcium chloride transformation for E. coli and calcium phosphate treatment or electroporation for mammalian cells. Cells transformed by the plasmids can be selected by resistance to antibiotics conferred by genes contained on the plasmids, such as the amp, gpt, neo and hyg genes.
Once expressed, the recombinant fusion proteins can be purified according to standard procedures of the art, including ammonium sulfate precipitation, affinity columns, column chromatography, gel electrophoresis and the like (see, generally, R. Scopes, Protein Purification, Springer-Verlag, New York (1982), Deutscher, Methods in Enzymology Vol. 182: Guide to Protein Purification, Academic Press, Inc. New York (1990)). Substantially pure compositions of at least about 90 to 95% homogeneity are preferred, and 98 to 99% or more homogeneity are most preferred for pharmaceutical uses. Once purified, partially or to homogeneity as desired, the polypeptides may then be used therapeutically.
One of skill in the art would recognize that after chemical synthesis, biological expression, or purification, the IL-13 receptor targeted fusion protein may possess a conformation substantially different than the native conformations of the constituent polypeptides. In this case, it may be necessary to denature and reduce the polypeptide and then to cause the polypeptide to re-fold into the preferred conformation. Methods of reducing and denaturing proteins and inducing re-folding are well known to those of skill in the art. (See, Debinski et al. J. Biol. Chem., 268: 14065-14070 (1993); Kreitman and Pastan, Bioconjug. Chem., 4: 581-585 (1993); and Buchner, et al., Anal. Biochem., 205: 263-270 (1992) which are incorporated herein by reference.) Debinski et al., for example, describe the denaturation and reduction of inclusion body proteins in guanidine-DTE. The protein is then refolded in a redox buffer containing oxidized glutathione and L-arginine.
One of skill would recognize that modifications can be made to the IL-13 receptor targeted fusion proteins without diminishing their biological activity. Some modifications may be made to facilitate the cloning, expression, or incorporation of the targeting molecule into a fusion protein. Such modifications are well known to those of skill in the art and include, for example, a methionine added at the amino terminus to provide an initiation site, or additional amino acids placed on either terminus to create conveniently located restriction sites or termination codons.
Identification of Target Cells
It was a surprising discovery of the present invention that tumor cells, overexpress IL-13 receptors. In particular, carcinoma tumor cells (e.g. renal carcinoma cells) overexpress IL-13 receptors at levels ranging from about 2100 sites/cell to greater than 150,000 sites per cell.
One of skill in the art will appreciate that identification of other cells that overexpress IL-13 receptors requires only routine screening using well-known methods. Typically this involves providing a labeled molecule that specifically binds to the IL-13 receptor. The cells in question are then contacted with this molecule and washed. Quantification of the amount of label remaining associated with the test cell provides a measure of the amount of IL-13 receptor (IL-13R) present on the surface of that cell.
In a preferred embodiment, IL-13 receptor may be quantified by measuring the binding of 125 I-labeled IL-13 (125 I-IL-13) to the cell in question. Details of such a binding assay are provided in Example 1.
Pharmaceutical Compositions
The chimeric molecules of this invention are useful for parenteral, topical, oral, or local administration, such as by aerosol or transdermally, for prophylactic and/or therapeutic treatment. The pharmaceutical compositions can be administered in a variety of unit dosage forms depending upon the method of administration. For example, unit dosage forms suitable for oral administration include powder, tablets, pills, capsules and lozenges. It is recognized that the fusion proteins and pharmaceutical compositions of this invention, when administered orally, must be protected from digestion. This is typically accomplished either by complexing the protein with a composition to render it resistant to acidic and enzymatic hydrolysis or by packaging the protein in an appropriately resistant carrier such as a liposome. Means of protecting proteins from digestion are well known in the art.
The pharmaceutical compositions of this invention are particularly useful for parenteral administration, such as intravenous administration or administration into a body cavity or lumen of an organ. The compositions for administration will commonly comprise a solution of the chimeric molecule dissolved in a pharmaceutically acceptable carrier, preferably an aqueous carrier. A variety of aqueous carriers can be used, e.g., buffered saline and the like. These solutions are sterile and generally free of undesirable matter. These compositions may be sterilized by conventional, well known sterilization techniques. The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, toxicity adjusting agents and the like, for example, sodium acetate, sodium chloride, potassium chloride, calcium chloride, sodium lactate and the like. The concentration of chimeric molecule in these formulations can vary widely, and will be selected primarily based on fluid volumes, viscosities, body weight and the like in accordance with the particular mode of administration selected and the patient's needs.
Thus, a typical pharmaceutical composition for intravenous administration would be about 0.1 to 10 mg per patient per day. Dosages from 0.1 up to about 100 mg per patient per day may be used, particularly when the drug is administered to a secluded site and not into the blood stream, such as into a body cavity or into a lumen of an organ. Actual methods for preparing parenterally administrable compositions will be known or apparent to those skilled in the art and are described in more detail in such publications as Remington's Pharmaceutical Science, 15th ed., Mack Publishing Company, Easton, Pa. (1980).
The compositions containing the present fusion proteins or a cocktail thereof (i.e., with other proteins) can be administered for therapeutic treatments. In therapeutic applications, compositions are administered to a patient suffering from a disease, in an amount sufficient to cure or at least partially arrest the disease and its complications. An amount adequate to accomplish this is defined as a "therapeutically effective dose." Amounts effective for this use will depend upon the severity of the disease and the general state of the patient's health.
Single or multiple administrations of the compositions may be administered depending on the dosage and frequency as required and tolerated by the patient. In any event, the composition should provide a sufficient quantity of the proteins of this invention to effectively treat the patient.
Among various uses of the cytotoxic fusion proteins of the present invention are included a variety of disease conditions caused by specific human cells that may be eliminated by the toxic action of the protein. One preferred application is the treatment of cancer, such as by the use of an IL-13 receptor targeting molecule (e.g. IL-13 or anti-IL-13R antibody) attached to a cytotoxin.
Where the chimeric molecule comprises an IL-13 receptor targeting molecule attached to a ligand, ligand portion of the molecule is chosen according to the intended use. Proteins on the membranes of T cells that may serve as targets for the ligand includes CD2 (T11), CD3, CD4 and CD8. Proteins found predominantly on B cells that might serve as targets include CD10 (CALLA antigen), CD19 and CD20. CD45 is a possible target that occurs broadly on lymphoid cells. These and other possible target lymphocyte target molecules for the chimeric molecules bearing a ligand effector are described in Leukocyte Typing III, A. J. McMichael, ed., Oxford University Press (1987). Those skilled in the art will realize ligand effectors may be chosen that bind to receptors expressed on still other types of cells as described above, for example, membrane glycoproteins or ligand or hormone receptors such as epidermal growth factor receptor and the like.
Diagnostic Kits
In another embodiment, this invention provides for kits for the treatment of tumors or for the detection of cells overexpressing IL-13 receptors. Kits will typically comprise a chimeric molecule of the present invention (e.g. IL-13-label, IL-13-cytotoxin, IL-13-ligand, etc.). In addition the kits will typically include instructional materials disclosing means of use of chimeric molecule (e.g. as a cytotoxin, for detection of tumor cells, to augment an immune response, etc.). The kits may also include additional components to facilitate the particular application for which the kit is designed. Thus, for example, where a kit contains a chimeric molecule in which the effector molecule is a detectable label, the kit may additionally contain means of detecting the label (e.g. enzyme substrates for enzymatic labels, filter sets to detect fluorescent labels, appropriate secondary labels such as a sheep anti-mouse-HRP, or the like). The kits may additionally include buffers and other reagents routinely used for the practice of a particular method. Such kits and appropriate contents are well known to those of skill in the art.
EXAMPLES
The following examples are offered to illustrate, but not to limit the claimed invention. All references cited in the foregoing discussion and the following examples are incorporated herein by reference.
Example 1 Identification of Cells that Overexpress IL-13
Recombinant human IL-4 and IL-13 were labeled with 125 I (Amersham Research Products, Arlington Heights, Ill., USA) by using the IODO-GEN reagent (Pierce, Rockford, Ill., USA) according to the manufacturer's instructions. The specific activity of the radiolabeled cytokines was estimated to range from 20-100 μCi/μg protein. For binding experiments, typically, 1×106 renal cell carcinoma (RCC) tumor cells were incubated at 4° C. for 2 hours with 125 I-IL-13 (100 pM) with or without increasing concentrations (up to 500 nM) of unlabeled IL-13. In some experiments, IL-13R expression was examined as previously described (Obiri et al. J. Clin. Invest., 91: 88-93 (1993))). The data were analyzed with the LIGAND program (Munson et al. Anal. Biochem., 107: 220-239 (1980)) to determine receptor number and binding affinity.
Four human renal cell carcinoma (RCC) cell lines (WS-RCC, HL-RCC, PM-RCC, and MA-RCC) bound 125 I-IL-13 specifically and the density of IL-13R varied from 2100 sites per cell in WS-RCC cells to 150,000 sites per cell in HL-RCC cells (Table 1). The represents an increase in IL-13 receptor expression ranging from 15 to about 500 fold as compared to normal immune cells. In contrast, IL-4 receptors overexpressed on cancers have been reported at concentrations as high as 4000 sites per cell. Scatchard analyses (Scatchard, Ann. N. Y. Acad. Sci., 51: 660-663 (1949)) revealed that only one affinity class of receptors was expressed on each cell line. The binding affinities (Kd) ranged between 100 pM to 400 pM in three RCC cell lines while HL-RCC cells expressed lower affinity receptors (Kd˜3 nM).
Although IL-13 responsiveness has previously been reported in human monocytes, B cells and pre-myeloid (TF-1) cells (see, e.g. de Waal Malefyt, et al. J. Immunol., 151: 6370-6381 (1993), de Waal Malefyt, et al. J. Immunol., 144: 629-633 (1993)), little was known about IL-13R structure or its binding characteristics in these, or any other cells. The present data show that freshly isolated human monocytes, EBV-transformed B cell line and TF-1 cell line express very few IL-13 binding sites (100-300/cell) compared to human RCC cells (Table 1). On the other hand, no binding of 125 I-IL-13 was observed on H9 T cells, LAK cells and resting or PHA activated PBL. This is compatible with the fact that IL-13 responsiveness has not been observed in T lymphocytes (Punnonen et al., Proc. Natl. Acad. Sci. USA, 90: 3730-3734 (1993).
              TABLE 1
______________________________________
Expression of IL-13 receptor by human cells
                IL-13 Binding
                Sites/cell.sup.a
                             Kd(nM)
Cell Types      Mean ± SD Mean ± SD
______________________________________
Renal Cell Carcinoma (RCC)
1. WS-RCC        2,090 ± 367 (5)
                             0.247 ± 0.12 (3).sup.b
2. MA-RCC        5,013 ± 1.347 (5)
                             0.128 ± 0.05 (2)
3. PM-RCC        26,500 ± 5.000 (2)
                             0.394 ± 0.26 (2)
4. HL-RCC       150,000 ± 15.00 (3)
                              3.1 ± 0.7 (2)
B Lymphocytes
1. DH (BBV-transformed B cell
                   303 ± 90 (4)
                               --.sup.d
line)
2. RAJI (Burkitt's lymphoma)
                UD.sup.c       --
Monocytes/Premyeloid cells.sup.e
1. Peripheral blood monocytes
                124            --
2. U937 (premonocytic
                UD             --
3. TF1.J61 (premyeloid)
                   130 ± 1 (2)
                               --
T Lymphocytes/LAK cells.sup.f
1. PHA-activated PBL
                <30            --
2. MOLT-4 (T-cell leukemia)
                UD             --
3. LAK cells    UD             --
______________________________________
 .sup.a IL13 binding sites/cell were determined as described in Example 1.
 .sup.b (n) = number of experiments used to calculate mean ± standard
 deviation.
 .sup.c UC = undetectable
 .sup.d The Kd could not be reliably calculated because of low binding of
 .sup.125 IIL-13
 .sup.e The peripheral blood derived monocytes (>90% purity) were isolated
 by ficollhypaque density gradient followed by ellutriation from a
 leukopac; obtained from normal donor.
 .sup.f LAK cells and activiated Tlymphocytes were generated by the cultur
 of donor PBLs (106/ml) with IL2 (500 Units/ml) for 3 days or PHA (10
 μg/ml) for 3-4 days respectively.
Example 2 IL-13 and IL-4 Bind to Different Receptors
Recently, it was proposed that the IL-2Rγc receptor subunit is associated with IL-13R (see, e.g., Russell et al. Science 262: 1880-1883 (1993); Kondo et al. Science, 262: 1874-1877 (1993); Noguchi et al. Science, 262: 1877-1880 (1993); Kondo et al. Science 263: 1453-1454 (1994); Giri et al. EMBO J. 13: 2822-2830 (1994))) and IL-13R may share a common component with IL-4R (Zurawski et al. EMBO J. 12: 2663-2670 (1993); Aversa et al. J. Exp. Med. 178: 2213-2218 (1993)). To directly address these possibilities, radio-ligand binding experiments were performed, as described in Example 1, on HL-RCC and WS-RCC cells using 125 I-IL-4 or 125 I-IL-13 in the presence or absence of excess of either cytokine.
Unlabeled IL-4 more efficiently inhibited 125 I-IL-4 from binding to RCC cells (84%, and 72% displacement of total binding in WS-RCC and HL-RCC, respectively) than IL-13 which also displaced 125 I-IL-4 binding to these cells (61% of total binding in WS-RCC and 51% in HL-RCC) under similar conditions. On the other hand, while 125 I-IL-13 binding was effectively displaced by IL-13 (about 85% of total in both cell types), it was only minimally displaced by IL-4 (12% of total displacement in WS-RCC, and 7% in HL-RCC). These results indicate that IL-4 and IL-13 both interact with each other's receptors, however, the interaction is not identical since IL-4 inhibition of 125 I-IL-13 binding was weak and IL-13 inhibition of I125 I-IL-4 binding was not complete. These results agree with previous observations in which IL-13 was found to compete with IL-4 binding on TF-1 cells (Zurawski et al., EMBO J. 12: 2663-2670 (1993)). However, in that report the converse experiment was not done. Here, the data show that even though IL-13 competed for IL-4 binding, IL-4 did not compete for IL-13 binding.
The competition by IL-13 for IL-4 binding sites on lymphoid MLA 144 cells and RAJI cell lines was also investigated. These cells were incubated with radiolabled IL-4 with or without excess unlabeled IL-4 or IL-13. Excess unlabeled IL-4 effectively displaced labeled 125 I-IL-4 bound to MLA 144 and RAJI cells, while excess IL-13 could not compete this binding. This observation is at variance to that seen with RCC cells in which IL-13 competed for IL-4 binding. The inability of IL-13 to compete for 125 I-IL-4 binding to MLA 144 is consistent with the observation that IL-13 did not bind to peripheral blood T (or MLA 144) cells.
Example 3 Subunit Structure of IL-13 and IL-4 Receptors
The subunit structure of IL-13R on RCC cells was investigated by crosslinking studies. Cells (5×106) were labeled with 125 I-IL-13 or 125 I-IL-4 in the presence or absence of excess IL-13 or IL-4 for 2 h at 4° C. The bound ligand was cross-linked to its receptor with disuccinimidyl suberate (DSS) (Pierce, Rockford, Ill., USA) at a final concentration of 2 mM for 30 min. Cells were lysed in a buffer containing 1% Triton X-100, 1 mM phenylmethylsulfonyl fluoride, 0.02 mM leupeptin, 5.0 μM trypsin inhibitor, 10 mM benzamidine HCl, 1 mM phenanthroline iodoacetamide, 50 mM amino caproic acid, 10 μg/ml pepstatin, and 10 μg/ml aprotinin. The cell lysates were cleared by boiling in buffer containing 2-mercaptoethanol and analyzed by electrophoresis through 8% SDS/polyacrylamide gel. The gel was subsequently dried and autoradiographed. In some experiments, the receptor/ligand complex was immunoprecipitated from the lysate overnight at 4° C. by incubating with protein A sepharose beads that had been pre-incubated with P7 anti hIL-4R or anti-γc antibody and analyzed as above.
The labeled 125 I-IL-13 cross-linked to one major protein on all four RCC cell lines and the complex migrated as a single broad band ranging between 68 and 80 kDa. A single band was also observed on human pre-myeloid TF-1.J61 cells only after much longer exposure of the gel. After subtracting the molecular mass of IL-13 (12 kDa), the size of IL-13 binding protein was estimated at 56 to 68 kDa. The 125 I-IL-13 cross-linked band was not observed when the crosslinking was performed in the presence of 200-fold molar excess of IL-13. In addition to the major band, a faint band of approximately 45 kDa was also observed in HL-RCC and PM-RCC but not on MA-RCC cells. This band appeared to be specifically associated with IL-13R because unlabeled IL-13 competed for the binding of 125 I-IL-13. This band could represent an IL-13R associated protein or a proteolytic fragment of the larger band. In contrast to the displacement of 125 I-IL-13 binding by unlabeled IL-13, an excess of unlabeled IL-4 did not prevent the appearance of IL-13R band in RCC cell lines. IL-13 on the other hand competed for 125 I-IL-4 binding to both major proteins on WS-RCC cells. It is of interest that 125 I-IL-13-cross-linked protein was slightly larger in size in TF-1.J61, WS-RCC, PM-RCC, and HL-RCC cell lines compared to that seen in MA-RCC. Post-translational modifications, such as glycosylation or phosphorylation, may account for this difference.
Example 4 Construction of an IL-13-PE Fusion Protein
Construction of a Plasmid Encoding IL-13-PE38QQR
To construct the chimeric toxin a coding region of the human interleukin 13 (hIL-13) gene (plasmid JFE14-SRα) (Minty et al., Nature, 362: 248 (1993), McKenzie et al. Proc. Natl. Acad. Sci. USA, 90: 3735 (1987) which are incorporated herein by reference) was fused to a gene encoding PE38QQR, a mutated form of PE, thereby producing a construct (phuIL-13-Tx) encoding the chimeric molecule. Specifically, a DNA encoding human IL-13 was PCR-amplified from plasmid JFE14-SRα. New sites were introduced for the restriction endonucleases NdeI and Hind III at the 5' and 3' ends of the hIL-13 gene, respectively by PCR using a sense primer that incorporated the NdeI site and an antisense primer that incorporated the HindIII site.
The NdeII/HindIII fragment containing encoding hIL-13 was subcloned into a vector obtained by digestion of plasmid pWDMH4-38QQR (Debinski et al. Int. J. Cancer 58: 744-748 (1994)) or plasmid pSGC242FdN1 (Debinski et al. Clin. Res. 42: 251A, (abstr.) (1994) with NdeI and HindIII, to produce plasmid phuIL-13-Tx. The 5' end of the gene fusion was sequenced and showed the correct DNA of hIL-13.
Human interleukin 4 (hIL-4) was cloned into an expression vector in a similar way to hIL-13 using plasmid pWDMH4 (Debinski et al. J. Biol. Chem. 268: 14065-14070 (1993)) as a template for PCR amplification. Recombinant proteins were expressed in E. coli BL21 (λDE3) under control of the T7 late promoter (Id.). In addition to the T7 bacteriophage late promoter, the plasmids also carried a T7 transcription terminator at the end of the open reading frame of the protein, an f1 origin of replication and gene for ampicillin resistance (Debinski et al. J. Clin. Invest. 90: 405-411 (1992)). The plasmids were amplified in E. coli (HB101 or DH5α high efficiency transformation) (BRL) and DNA was extracted using Qiagen kits (Chatsworth, Calif., USA).
Expression and purification of recombinant proteins
E. coli BL21 (λDE3) cells were transformed with plasmids of interest and cultured in 1.0 liter of Super broth. Expressed recombinant human IL-13 and human IL-13-PE38QQR were localized in inclusion bodies. The recombinant proteins were isolated from the inclusion bodies as described by Debinski et al., J. Biol. Chem. 268: 14065-14070 (1993), which is incorporated herein by reference. After dialysis, the renatured protein of human IL-13-PE38QQR was purified on Q-Sepharose Fast Flow and by size exclusion chromatography on Sephacryl S-200HR (Pharmacia, Piscataway, N.J., USA) The initial step of hIL-13 or hIL-4 purification was conducted on SP-Sepharose Fast Flow (Pharmacia).
Protein concentration was determined by the Bradford assay (Pierce "Plus", Rockford, Ill. USA) using BSA as a standard.
Human IL-13 and IL-13-PE38QQR were expressed at high levels in bacteria as seen in SDS-PAGE analysis of the total cell extract. After initial purification on SP-Sepharose (hIL-13) or Q-Sepharose (hIL-13-PE38QQR) the renatured recombinant proteins were applied onto a Sephacryl S-200 HR Pharmacia column. Human IL-13 and hIL-13-PE38QQR appeared as single entities demonstrating the very high purity of the final products. The chimeric toxin migrated within somewhat lower than expected for 50 kDa protein Mr range which may be related to the hydrophobicity of the molecule. The biologic activity of the rhIL-13 was exactly the same as commercially obtained hIL-13.
Example 5 The Activity of an IL13-PE Fusion Protein on Human Carcinoma Cells
Cytotoxic Activity of hIL-13-PE38QQR
The cytotoxic activity of chimeric toxins, such as hIL-13-PE38QQR, were tested by measuring inhibition of protein synthesis. Protein synthesis was assayed by plating about 1×104 cells per in a 24-well tissue culture plate in 1 ml of medium. Various concentrations of the chimeric toxins were added 20-28 h following cell plating. After 20 h incubation with chimeric toxins, 3 H!-leucine was added to cells for 4 h, and the cell-associated radioactivity was measured. For blocking studies, rhIL-2, 4 or 13 was added to cells for 30 min before the chimeric toxin addition. Data were obtained from the average of duplicates and the assays were repeated several times.
Several established cancer cell lines were tested to determine if hIL-13-PE38QQR is cytotoxic to them. In particular, cancers derived from colon, skin and stomach were examined. The cancer cells were sensitive to hIL-13-PE38QQR with ID50 s ranging from less than 1 ng/ml to 300 ng/ml (20 pM to 6.0 nM) (ID50 indicates the concentration of the chimeric toxin at which the protein synthesis fell by 50% when compared to the sham-treated cells). A colon adenocarcinoma cell line, Colo201, was very responsive with an IC50 of 1 ng/ml. A431 epidermoid carcinoma cells were also very sensitive to the action of hIL-13-toxin; the ID50 for hIL-13-PE38QQR ranged from 6 to 10 ng/ml. A gastric carcinoma CRL1739 cell line responded moderately to the hIL-13-toxin with an ID50 of 50 ng/ml. Another colon carcinoma cell line, Colo205, had a poorer response with an ID50 of 300 ng/ml.
The cytotoxic action of hIL-13-PE38QQR was specific as it was blocked by a 10-fold excess of hIL-13 on all cells. These data suggest that a spectrum of human cancer cells possess hIL-13 binding sites and such cells are sensitive to hIL-13-PE38QQR chimeric toxin.
Because the hIL-13R has been suggested to share the λc subunit of the IL-2R (Russell et al. Science 262: 1880-1883 (1993)), the specificity of hIL-13-PE38QQR action on A431 and CRL1739 cells, the two cell lines with different sensitivities to the chimeric toxin was further explored. The cells were treated with hIL-13-PE38QQR with or without rhIL-2 at a concentration of 1.0 μg/ml or 10 μg/ml. The rhIL-2 did not have any blocking action on hIL-13-PE38QQR on the two cell lines, even at 10,000 fold molar excess over the chimeric toxin. These results indicate that the cell killing by the hIL-13-toxin is independent of the presence of hIL-2.
IL-4, unlike IL-2, blocks the action of IL-13-PE38QOR
Native hIL-4 was added to cells which were then treated with hIL-13-PE38QQR. Unexpectedly, it was found that hIL-4 inhibited the cytotoxic activity of the hIL-13-toxin. This phenomenon was seen on all the tested cell lines, including Colo201, A431 and CRL1739. To investigate the possibility that hIL-13 and hIL-4 may compete for the same binding site, the cells were also treated with the hIL-4-based recombinant toxin, hIL-4-PE38QQR (Debinski et al. Int. J. Cancer 8: 744-748 (1994)). The cytotoxic action of hIL-4-PE38QQR had already been shown to be blocked by an excess of hIL-4 but not of hIL-2 (Id.). In the present experiment hIL-13 potently blocked the cytotoxic activity of hIL-4-PE38QQR. Also, the action of another hIL-4-based chimeric toxin, hIL-4-PE4E (Debinski et al. J. Biol. Chem. 268: 14065-14070 (1993)), was blocked by an excess of hIL-13 on Colo201 and A431 cells. Thus, the cytotoxicity of hIL-13-PE38QQR is blocked by an excess of hIL-13 or hIL-4, and the cytotoxic action of hIL-4-PE38QQR is also blocked by the same two growth factors. However, IL-2 does not block the action of either chimeric toxin. These results strongly suggest that hIL-4 and hIL-13 have affinities for a common binding site.
This conclusion was supported by the observation of one cytokine blocking the effect of a mixture of the two chimeric toxins. When A431 cells were incubated with both hIL-3- and hIL-4-PE38QQR chimeric toxins concomitantly the cytotoxic action was preserved and additive effect was observed as expected. An excess of hIL-13 efficiently blocked the action of a mixture of the two chimeric toxins. Moreover, neither hIL-13 nor hIL-4 blocked cell killing by another mixture composed of hIL-13-PE38QQR and TGFα-PE40, a chimeric toxin which targets the EGFR (TGFα-based chimeric toxin, TGFα-PE40) (Siegall et al. FASEB J. 3, 2647-2652 (1992)). The same was observed on Colo201 cells.
Reciprocal Blocking of Chimeric Toxins by IL-13 and IL-4 is due to competition for binding sites
The binding ability of human IL-13 was compared to human IL-4-PE38QQR in competitive binding assays. Recombinant hIL-4-PE38QQR was labeled with 125 I using the lactoperoxidase method as described by Debinski et al., J. Clin. Invest. 90, 405-411 (1992). Binding assays were performed by a standard saturation and displacement curves analysis. A431 epidermoid carcinoma cells were seeded at 105 cells per well in a 24-well tissue culture plates at 24 h before the experiment. The plates were placed on ice and cells were washed with ice-cold PBS without Ca++, Mg++ in 0.2% BSA, as described (Id.). Increasing concentrations of hIL-13 or hIL-4-PE38QQR were added to cells and incubated 30 min prior to the addition of fixed amount of 125 I-hIL-4-PE38QQR (specific activity 6.2 μCi/μg protein) for 2 to 3 h. After incubation, the cells were washed twice and lysed with 0.1N NaOH, and the radioactivity was counted in a γ-counter.
Human IL-4-PE38QQR competed for the binding of 125 I-hIL-4-PE38QQR to A431 cells with an apparent ID50 of 4×10-8 M. In addition, hIL-13 also competed for the 125 I-hIL-4-PE38QQR binding site with a comparable potency to that exhibited by the chimeric protein. More extensive binding studies have shown that hIL-13 also competes for hIL-4 binding sites on human renal carcinoma cell lines.
The possibility of an influence of hIL-13 or hIL-4 on the process of receptor-mediated endocytosis and post-binding PE cellular toxicity steps was excluded by adding to cells: (i) native PE (PE binds to the α2 -macroglobulin receptor), (ii) TGFα-PE40, and (iii) a recombinant immunotoxin C242rF(ab)-PE38QQR (Debinski et al. Clin. Res. 42, 251A, (Abstr.) (1994)). C242rF(ab)-PE38QQR binds a tumor-associated antigen that is a sialylated glycoprotein (Debinski et al. J. Clin. Invest. 90: 405-411 (1992)). The expected cytotoxic actions of these recombinant toxins were observed and neither hIL-13 nor hIL-4 blocked these actions on A431 and Colo205 cells.
hIL-4 and hIL-13 compete for a common binding site on carcinoma cells but evoke different biological effects
Even though hIL-13 and hIL-4 compete for a common binding site, they induce different cellular effects. Protein synthesis was inhibited in A431 epidermoid carcinoma cells in a dose-dependent manner by hIL-4 alone, or by a ADP-ribosylation deficient chimeric toxin containing hIL-4 (Debinski et al. Int. J. Cancer 58: 744-748 (1994)). This effect of hIL-4 or enzymatically deficient chimeric toxin can be best seen with a prolonged time of incubation (≧24 h) and requires concentrations of hIL-4 many fold higher than that of the active chimeric toxin in order to cause a substantial decrease in tritium incorporation. However, when A431 cells were treated with various concentrations of hIL-13, no inhibition (or stimulation) of protein synthesis was observed, even at concentrations as high as 10 μg/ml of hIL-13 for a 72 h incubation. The same lack of response to hIL-13 was found on renal cell carcinoma cells PM-RCC. Thus, while hIL-13 and hIL-4 may possess a common binding site, they appear to transduce differently in carcinoma cells expressing this common site, such as A431 and PM-RCC cells.
The above examples are provided to illustrate the invention but not to limit its scope. Other variants of the invention will be readily apparent to one of ordinary skill in the art and are encompassed by the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference.
__________________________________________________________________________
#             SEQUENCE LISTING
- (1) GENERAL INFORMATION:
-    (iii) NUMBER OF SEQUENCES: 5
- (2) INFORMATION FOR SEQ ID NO:1:
-      (i) SEQUENCE CHARACTERISTICS:
#acids    (A) LENGTH: 5 amino
          (B) TYPE: amino acid
          (C) STRANDEDNESS:
          (D) TOPOLOGY: linear
-     (ii) MOLECULE TYPE: peptide
#ID NO:1: (xi) SEQUENCE DESCRIPTION: SEQ
-      Gly Gly Gly Gly Ser
#  5 1
- (2) INFORMATION FOR SEQ ID NO:2:
-      (i) SEQUENCE CHARACTERISTICS:
#acids    (A) LENGTH: 5 amino
          (B) TYPE: amino acid
          (C) STRANDEDNESS:
          (D) TOPOLOGY: linear
-     (ii) MOLECULE TYPE: peptide
#ID NO:2: (xi) SEQUENCE DESCRIPTION: SEQ
-      Arg Glu Asp Leu Lys
#  5 1
- (2) INFORMATION FOR SEQ ID NO:3:
-      (i) SEQUENCE CHARACTERISTICS:
#acids    (A) LENGTH: 4 amino
          (B) TYPE: amino acid
          (C) STRANDEDNESS:
          (D) TOPOLOGY: linear
-     (ii) MOLECULE TYPE: peptide
#ID NO:3: (xi) SEQUENCE DESCRIPTION: SEQ
-      Arg Glu Asp Leu
     1
- (2) INFORMATION FOR SEQ ID NO:4:
-      (i) SEQUENCE CHARACTERISTICS:
#acids    (A) LENGTH: 4 amino
          (B) TYPE: amino acid
          (C) STRANDEDNESS:
          (D) TOPOLOGY: linear
-     (ii) MOLECULE TYPE: peptide
#ID NO:4: (xi) SEQUENCE DESCRIPTION: SEQ
-      Arg Asp Glu Leu
     1
- (2) INFORMATION FOR SEQ ID NO:5:
-      (i) SEQUENCE CHARACTERISTICS:
#acids    (A) LENGTH: 4 amino
          (B) TYPE: amino acid
          (C) STRANDEDNESS:
          (D) TOPOLOGY: linear
-     (ii) MOLECULE TYPE: peptide
#ID NO:5: (xi) SEQUENCE DESCRIPTION: SEQ
-      Lys Asp Glu Leu
__________________________________________________________________________

Claims (14)

What is claimed is:
1. A chimeric molecule that specifically binds a tumor cell bearing an IL-13 receptor, said chimeric molecule comprising a cytotoxin attached to a targeting molecule that is an IL-13 molecule or an antibody that specifically binds an IL-13 receptor.
2. The chimeric molecule of claim 1, wherein said targeting molecule is human IL-13.
3. The chimeric molecule of claim 1, wherein said cytotoxin is selected from the group consisting of Pseudomonas exotoxin, ricin, abrin and Diphtheria toxin.
4. The chimeric molecule of claim 3, wherein chimeric molecule is a single-chain fusion protein.
5. The chimeric molecule of claim 4, wherein said cytotoxin is a Pseudomonas exotoxin.
6. The chimeric molecule of claim 5, wherein said Pseudomonas exotoxin is PE38QQR.
7. A chimeric molecule that specifically binds a tumor cell bearing an IL-13 receptor, said chimeric molecule comprising an effector molecule attached to an antibody that specifically binds an IL-13 receptor,
wherein said effector molecule is selected from the group consisting of a cytotoxin, a label, a radionuclide and a liposome, wherein the liposome contains a cytotoxin, a label, or a radionuclide.
8. A composition comprising a pharmacologically acceptable carrier and a chimeric molecule that specifically binds a tumor cell bearing an IL-13 receptor, said chimeric molecule comprising:
an effector molecule attached to
a targeting molecule that is an IL-13 molecule or an antibody that specifically binds an IL-13 receptor,
wherein said effector molecule is selected from the group consisting of a cytotoxin, a label, a radionuclide and a liposome, wherein the liposome contains a cytotoxin, a label, or a radionuclide.
9. The composition of claim 8, wherein said targeting molecule is IL-13.
10. The composition of claim 9, wherein chimeric molecule is a single-chain fusion protein.
11. The composition of claim 10, wherein said effector molecule is a Pseudomonas exotoxin.
12. The composition of claim 11, wherein said Pseudomonas exotoxin is PE38QQR.
13. A chimeric molecule that specifically binds a tumor cell bearing an IL-13 receptor, said chimeric molecule comprising an effector molecule attached to targeting molecule comprising an antibody that specifically binds an IL-13 receptor, wherein said effector molecule is linked to said targeting molecule by a linker consisting of a ligand.
14. A composition comprising a pharmacologically acceptable carrier and a chimeric molecule that specifically binds a tumor cell bearing an IL-13 receptor, said chimeric molecule comprising:
an effector molecule attached to
a targeting molecule that is an IL-13 molecule or an antibody that specifically binds an IL-13 receptor, wherein said effector molecule is linked to said targeting molecule by a linker consisting of a ligand.
US08/821,840 1995-03-15 1997-03-21 IL-13 receptor specific chimeric proteins Expired - Lifetime US5919456A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US08/821,840 US5919456A (en) 1995-03-15 1997-03-21 IL-13 receptor specific chimeric proteins

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/404,685 US5614191A (en) 1995-03-15 1995-03-15 IL-13 receptor specific chimeric proteins and uses thereof
US08/821,840 US5919456A (en) 1995-03-15 1997-03-21 IL-13 receptor specific chimeric proteins

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US08/404,685 Division US5614191A (en) 1995-03-15 1995-03-15 IL-13 receptor specific chimeric proteins and uses thereof

Publications (1)

Publication Number Publication Date
US5919456A true US5919456A (en) 1999-07-06

Family

ID=23600619

Family Applications (2)

Application Number Title Priority Date Filing Date
US08/404,685 Expired - Lifetime US5614191A (en) 1995-03-15 1995-03-15 IL-13 receptor specific chimeric proteins and uses thereof
US08/821,840 Expired - Lifetime US5919456A (en) 1995-03-15 1997-03-21 IL-13 receptor specific chimeric proteins

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US08/404,685 Expired - Lifetime US5614191A (en) 1995-03-15 1995-03-15 IL-13 receptor specific chimeric proteins and uses thereof

Country Status (9)

Country Link
US (2) US5614191A (en)
EP (1) EP1007696B1 (en)
JP (1) JP4202417B2 (en)
AT (1) ATE417931T1 (en)
AU (1) AU714541B2 (en)
CA (1) CA2215122A1 (en)
DE (1) DE69637781D1 (en)
ES (1) ES2319827T3 (en)
WO (1) WO1996029417A1 (en)

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001034645A2 (en) * 1999-11-11 2001-05-17 The Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services Mutated il-13 molecules and their uses
US6346247B1 (en) 1999-10-28 2002-02-12 Promega Corporation Prevention and treatment of autoimmune disease with luminally administered polyclonal antibodies
US6518061B1 (en) 1995-03-15 2003-02-11 The United States Of America As Represented By The Department Of Health And Human Services IL-13 receptor specific chimeric proteins and uses thereof
US6576232B1 (en) * 1998-04-03 2003-06-10 The Penn State Research Foundation IL13 mutants
US20030129132A1 (en) * 1998-02-17 2003-07-10 The Government Of The Usa As Represented By The Secretary Of The Dept. Of Health & Human Services IL-13 receptor specific chimeric proteins & uses thereof
US20030144229A1 (en) * 2000-01-14 2003-07-31 The Government Of The U.S.A., The Secretary Of The Department Of Health And Human Services Multiple CpG oligodeoxynucleotides and their use to induce an immune response
US20030194391A1 (en) * 2001-03-03 2003-10-16 Claire Ashman Vaccine
US20030215421A1 (en) * 1999-07-21 2003-11-20 Mcdonald John R. Methods and compositions for treating secondary tissue damage and other inflammatory conditions and disorders
US6703360B2 (en) 2000-04-07 2004-03-09 Heska Corporation Compositions and methods related to canine IgG and canine IL-13 receptors
US20040105872A1 (en) * 2002-09-18 2004-06-03 The Government Of The Usa As Represented By The Secretary Of The Dept. Of Health & Human Services Method of treating and preventing infections in immunocompromised subjects with immunostimulatory CpG oligonucleotides
US20050002918A1 (en) * 2001-11-09 2005-01-06 Neopharm, Inc. Selective treatment of IL-13 expressing tumors
US20050026245A1 (en) * 2001-12-20 2005-02-03 Klinman Dennis M. Use of cpg oligodeoxynucleotides to induce angiogenesis
US20050058645A1 (en) * 2002-03-22 2005-03-17 Amrad Operations Pty. Ltd. Monoclonal antibody against Interleukin-13 receptor alpha 1 (IL-13Ralpha1)
US20050142105A1 (en) * 2001-12-04 2005-06-30 Puri Raj K. Chimeric molecule for the treatment of th2-like cytokine mediated disorders
US20050209184A1 (en) * 1999-04-12 2005-09-22 The Govt. Of The Usa As Represented By The Secretary Of The Dept Of Health And Human Services Oligodeoxynucleotide and its use to induce an immune response
US20050214355A1 (en) * 2001-07-27 2005-09-29 Klinman Dennis M Washing and chilling apparatus and method
US20060099652A1 (en) * 2003-03-26 2006-05-11 Neopharm, Inc. IL 13 receptor alpha 2 antibody and methods of use
US20060140948A1 (en) * 2004-11-17 2006-06-29 Ian Foltz Fully human monoclonal antibodies to IL-13
WO2006113614A2 (en) 2005-04-15 2006-10-26 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Service National Institutes Of Health Methods of treating and preventing inflammatory bowel disease involving il-13 and nkt cells
US7157418B1 (en) 1998-07-22 2007-01-02 Osprey Pharmaceuticals, Ltd. Methods and compositions for treating secondary tissue damage and other inflammatory conditions and disorders
US7354909B2 (en) 2001-08-14 2008-04-08 The United States Of America As Represented By Secretary Of The Department Of Health And Human Services Method for rapid generation of mature dendritic cells
WO2008049098A3 (en) * 2006-10-19 2009-04-09 Merck & Co Inc ANTIBODY ANTAGONISTS OF INTERLEUKIN-13 RECEPTOR α1
US8343461B2 (en) 2006-03-08 2013-01-01 Wake Forest University Health Sciences Molecular signature of cancer
US8362207B2 (en) 2010-04-16 2013-01-29 Wake Forest University Health Sciences Multi-level specific targeting of cancer cells with IL-13
US8466116B2 (en) 2001-12-20 2013-06-18 The Unites States Of America As Represented By The Secretary Of The Department Of Health And Human Services Use of CpG oligodeoxynucleotides to induce epithelial cell growth
WO2015070210A1 (en) 2013-11-11 2015-05-14 Wake Forest University Health Sciences Epha3 and multi-valent targeting of tumors
EP2946786A1 (en) 2006-03-08 2015-11-25 Wake Forest University Health Sciences Soluble monomeric Ephrin A1
US9296785B2 (en) 2009-04-17 2016-03-29 Wake Forest University Health Sciences IL-13 receptor binding peptides
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria

Families Citing this family (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20010053371A1 (en) * 1999-01-07 2001-12-20 Waldemar Debinski Method for diagnosing, imaging, and treating tumors using restrictive receptor for interleukin 13
US5614191A (en) * 1995-03-15 1997-03-25 The United States Of America As Represented By The Department Of Health And Human Services IL-13 receptor specific chimeric proteins and uses thereof
US6428788B1 (en) 1995-03-15 2002-08-06 Penn State University Compositions and methods for specifically targeting tumors
US6803188B1 (en) * 1996-01-31 2004-10-12 The Regents Of The University Of California Tandem fluorescent protein constructs
US6900304B2 (en) 1996-01-31 2005-05-31 The Regents Of The University Of California Emission ratiometric indicators of phosphorylation
CA2202103C (en) * 1996-04-11 2007-01-09 Toshiaki Tagawa Method for preparing closed vesicles
US5925558A (en) * 1996-07-16 1999-07-20 The Regents Of The University Of California Assays for protein kinases using fluorescent protein substrates
US5912137A (en) * 1996-07-16 1999-06-15 The Regents Of The University Of California Assays for protein kinases using fluorescent
AU737910B2 (en) * 1997-01-31 2001-09-06 Regents Of The University Of California, The Chimeric antibody fusion proteins for the recruitment and stimulation of an antitumor immune response
US6197928B1 (en) 1997-03-14 2001-03-06 The Regents Of The University Of California Fluorescent protein sensors for detection of analytes
US5998204A (en) * 1997-03-14 1999-12-07 The Regents Of The University Of California Fluorescent protein sensors for detection of analytes
DE19735105A1 (en) * 1997-08-13 1999-03-04 Univ Albert Ludwigs Freiburg New fusion protein
GB9725126D0 (en) * 1997-11-27 1998-01-28 Svanborg Catharina Therapeutic agents
US6296843B1 (en) * 1998-04-03 2001-10-02 The Penn State Research Foundation Mutagenized IL 13-based chimeric molecules
AU2007201749B2 (en) * 1998-07-22 2008-05-29 Osprey Pharmaceuticals Usa, Inc. Cytotoxic conjugates comprising a chemokine receptor targeting agent
US6495664B1 (en) * 1998-07-24 2002-12-17 Aurora Biosciences Corporation Fluorescent protein sensors of post-translational modifications
US6410255B1 (en) 1999-05-05 2002-06-25 Aurora Biosciences Corporation Optical probes and assays
EP1222212A4 (en) * 1999-10-06 2005-05-04 Penn State Res Found Il13 mutants
CA2398136A1 (en) * 2000-02-08 2001-08-16 The Penn State Research Foundation Immunotherapy using interleukin 13 receptor subunit alpha 2
EP1315742B1 (en) * 2000-08-30 2010-06-30 The Penn State Research Foundation Amino acid substitution mutants of interleukin 13
WO2002017968A2 (en) * 2000-08-31 2002-03-07 The Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services Sensitization of cancer cells to immunoconjugate-induced cell death by transfection with il-13 receptor alpha chain-2
DE10045592A1 (en) * 2000-09-15 2002-03-28 Klaus Pfizenmaier An antibody-TNF-TNF inhibitor fusion protein (TNF selectokine) as a target-specific procytokine for tumor therapy
GB0112687D0 (en) * 2001-05-24 2001-07-18 Microbiological Res Authority Pharmaceutical use of secreted bacterial effector proteins
IL159215A0 (en) * 2001-06-07 2004-06-01 Wyeth Corp A solution of interleukin-13 and methods utilizing the same
JP3829252B2 (en) * 2001-06-08 2006-10-04 独立行政法人理化学研究所 Fluorescent protein
EP1490109A4 (en) * 2002-03-19 2006-04-19 Penn State Res Found Egfr ligands and methods of use
CA2480008C (en) * 2002-03-22 2015-10-06 The Penn State Research Foundation Use of il-13 receptor subunit alpha 2 as an anti-cancer vaccine
WO2003086451A1 (en) * 2002-04-05 2003-10-23 Centocor, Inc. Asthma-related anti-il-13 immunoglobulin derived proteins, compositions, methods and uses
EP1498137A1 (en) * 2002-04-25 2005-01-19 Chugai Seiyaku Kabushiki Kaisha Remedy for lung cancer
GB0210464D0 (en) 2002-05-08 2002-06-12 Svanborg Catharina Therapeutic treatment
GB0211658D0 (en) * 2002-05-22 2002-07-03 Svanborg Catharina Novel therapies and methods of screening for therapeutic compounds
DE60239566D1 (en) * 2002-06-14 2011-05-05 Brigham And Women S Hospital Boston PROCESS FOR TREATMENT AND PREVENTION OF KOLITIS IN WHICH IL-13 AND NK-T CELLS ARE INVOLVED
EP1628679A2 (en) * 2003-05-23 2006-03-01 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumors of glial origin
CA2445420A1 (en) * 2003-07-29 2005-01-29 Invitrogen Corporation Kinase and phosphatase assays
US7619059B2 (en) * 2003-07-29 2009-11-17 Life Technologies Corporation Bimolecular optical probes
US7727752B2 (en) * 2003-07-29 2010-06-01 Life Technologies Corporation Kinase and phosphatase assays
DE602004025592D1 (en) * 2003-09-12 2010-04-01 Life Technologies Corp Applications of resonance energy transfer between terbium and GFP
EP1687016A4 (en) * 2003-11-25 2009-07-29 Anjin Corp Diphtheria toxin variant
WO2005079755A2 (en) * 2004-02-12 2005-09-01 Nektar Therapeutics Interleukin-13 antagonist powders, spray-dried particles, and methods
WO2005091853A2 (en) * 2004-02-27 2005-10-06 Centocor, Inc. Methods and compositions for treating il-13 related pathologies
TWI306862B (en) 2005-01-03 2009-03-01 Hoffmann La Roche Antibodies against il-13 receptor alpha 1 and uses thereof
US20070160658A1 (en) * 2005-10-20 2007-07-12 The Penn State Research Foundation Delivery system for diagnostic and therapeutic agents
JP2010190572A (en) * 2007-06-01 2010-09-02 Sapporo Medical Univ Antibody directed against il13ra2, and diagnostic/therapeutic agent comprising the antibody
AU2008337904B2 (en) 2007-12-15 2014-02-13 F. Hoffmann-La Roche Ag Distinguishing assay
SI2859015T1 (en) 2012-06-08 2018-12-31 Alkermes Pharma Ireland Limited Ligands modified by circular permutation as agonists and antagonists
CN105378480B (en) 2013-07-04 2018-06-12 豪夫迈·罗氏有限公司 Detect the AF panel immunoassays of anti-drug antibodies in blood serum sample
WO2017075537A1 (en) 2015-10-30 2017-05-04 Aleta Biotherapeutics Inc. Compositions and methods for treatment of cancer
CN108472365A (en) * 2015-10-30 2018-08-31 艾丽塔生物治疗剂公司 Composition and method for tumour transduction
WO2018106931A1 (en) 2016-12-07 2018-06-14 Progenity Inc. Gastrointestinal tract detection methods, devices and systems
WO2018183932A1 (en) 2017-03-30 2018-10-04 Progenity Inc. Treatment of a disease of the gastrointestinal tract with a il-13 inhibitor
EP3883635A1 (en) 2018-11-19 2021-09-29 Progenity, Inc. Methods and devices for treating a disease with biotherapeutics
US11707610B2 (en) 2019-12-13 2023-07-25 Biora Therapeutics, Inc. Ingestible device for delivery of therapeutic agent to the gastrointestinal tract

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5082927A (en) * 1986-09-24 1992-01-21 The United States Of America As Represented By The Department Of Health And Human Services Selectively cytotoxic IL-4-PE40 fusion protein
WO1994004680A1 (en) * 1992-08-21 1994-03-03 Schering Corporation Human interleukin-13
US5596072A (en) * 1992-08-21 1997-01-21 Schering Corporation Method of refolding human IL-13
US5614191A (en) * 1995-03-15 1997-03-25 The United States Of America As Represented By The Department Of Health And Human Services IL-13 receptor specific chimeric proteins and uses thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5082927A (en) * 1986-09-24 1992-01-21 The United States Of America As Represented By The Department Of Health And Human Services Selectively cytotoxic IL-4-PE40 fusion protein
WO1994004680A1 (en) * 1992-08-21 1994-03-03 Schering Corporation Human interleukin-13
US5596072A (en) * 1992-08-21 1997-01-21 Schering Corporation Method of refolding human IL-13
US5614191A (en) * 1995-03-15 1997-03-25 The United States Of America As Represented By The Department Of Health And Human Services IL-13 receptor specific chimeric proteins and uses thereof

Non-Patent Citations (25)

* Cited by examiner, † Cited by third party
Title
Chaudhary et al., Nature, 339: 394 397 (1989). *
Chaudhary et al., Nature, 339: 394-397 (1989).
Chester et al., Tibtec 13: vol. 13, 294 300 (1995). *
Chester et al., Tibtec 13: vol. 13, 294-300 (1995).
Debinski et al., Biocanjugate Chem, vol. 5, 40 46 (1994). *
Debinski et al., Biocanjugate Chem, vol. 5, 40-46 (1994).
Debinski et al., J. Biol Chem, vol. 268, 19:14065 (1993). *
EMBO Journal, vol. 12, No. 7, 1993, Eynsham, Oxford GB, pp. 2663 2670, XP002011860 S. M. Zurawski et al: Receptors for Interleukin 13 and Interleukin 4 are complex and share a novel component that functions in signal transduction . *
EMBO Journal, vol. 12, No. 7, 1993, Eynsham, Oxford GB, pp. 2663-2670, XP002011860 S. M. Zurawski et al: "Receptors for Interleukin-13 and Interleukin-4 are complex and share a novel component that functions in signal transduction".
Gottstein et al., Annab of Oncology, vol. 5 Supplement 1 S97 103 (1994). *
Gottstein et al., Annab of Oncology, vol. 5 Supplement 1 S97-103 (1994).
Journal of Biological Chemistry, vol. 270, No. 28, 14 Jul. 1995, MD US, pp. 16775 16780, XP002011861 W. Debinski et al: A novel chimeric protein composed of Interleukin 13 and Pseudomonas Exotoxin is highly cytotoxic to human carcinoma cells expressing receptors for Interleukin 13 and Interleukin 4 . *
Journal of Biological Chemistry, vol. 270, No. 28, 14 Jul. 1995, MD US, pp. 16775-16780, XP002011861 W. Debinski et al: "A novel chimeric protein composed of Interleukin-13 and Pseudomonas Exotoxin is highly cytotoxic to human carcinoma cells expressing receptors for Interleukin-13 and Interleukin-4".
McKenzie et al. Proc. Natl. Acad. Sci. USA, 90: 3735 (1993). *
Minty et al., Nature, 362: 248 (1993). *
Obiri et al., J. Biol. Chem., vol. 270, 15:8797 (1995). *
Pastan et al., Ann. Rev. Biochem., 61: 331 354 (1992). *
Pastan et al., Ann. Rev. Biochem., 61: 331-354 (1992).
Paul Fundamental Immunology Third Edition 784 789, 1993. *
Paul Fundamental Immunology Third Edition 784-789, 1993.
Puri et al Blood vol 87 No. 10 4333 4339, May 1996. *
Puri et al Blood vol 87 No. 10 4333-4339, May 1996.
Thrush et al., Ann Rev Immunol., vol. 14, 49 71 (1996). *
Thrush et al., Ann Rev Immunol., vol. 14, 49-71 (1996).
Vita et al., Journ. of biol. Chem., 270:3512 (1995). *

Cited By (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6518061B1 (en) 1995-03-15 2003-02-11 The United States Of America As Represented By The Department Of Health And Human Services IL-13 receptor specific chimeric proteins and uses thereof
US20030129132A1 (en) * 1998-02-17 2003-07-10 The Government Of The Usa As Represented By The Secretary Of The Dept. Of Health & Human Services IL-13 receptor specific chimeric proteins & uses thereof
US6576232B1 (en) * 1998-04-03 2003-06-10 The Penn State Research Foundation IL13 mutants
US20110160126A1 (en) * 1998-07-22 2011-06-30 Mcdonald John R Methods and compositions for treating secondary tissue damage and other inflammatory conditions and disorders
US7192736B2 (en) 1998-07-22 2007-03-20 Osprey Pharmaceuticals, Ltd. Nucleic acid molecules encoding cytotoxic conjugates that contain a chemokine receptor targeting agent
US7166702B1 (en) 1998-07-22 2007-01-23 Osprey Pharmaceuticals, Ltd. Cytotoxic conjugates comprising a chemokine receptor targeting agent
US7157418B1 (en) 1998-07-22 2007-01-02 Osprey Pharmaceuticals, Ltd. Methods and compositions for treating secondary tissue damage and other inflammatory conditions and disorders
US6977245B2 (en) 1999-04-12 2005-12-20 The United States Of America As Represented By The Department Of Health And Human Services Oligodeoxynucleotide and its use to induce an immune response
US20050209184A1 (en) * 1999-04-12 2005-09-22 The Govt. Of The Usa As Represented By The Secretary Of The Dept Of Health And Human Services Oligodeoxynucleotide and its use to induce an immune response
US7960356B2 (en) 1999-04-12 2011-06-14 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Oligodeoxynucleotide and its use to induce an immune response
US8227446B2 (en) 1999-04-12 2012-07-24 The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services Oligodeoxynucleotide and its use to induce an immune response
US20030215421A1 (en) * 1999-07-21 2003-11-20 Mcdonald John R. Methods and compositions for treating secondary tissue damage and other inflammatory conditions and disorders
EP1949914A1 (en) 1999-10-28 2008-07-30 Promega Corporation Prevention and treatment of autoimmune disease with luminally administered polyclonal antibodies
US6346247B1 (en) 1999-10-28 2002-02-12 Promega Corporation Prevention and treatment of autoimmune disease with luminally administered polyclonal antibodies
WO2001034645A2 (en) * 1999-11-11 2001-05-17 The Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services Mutated il-13 molecules and their uses
WO2001034645A3 (en) * 1999-11-11 2002-03-07 Us Health Mutated il-13 molecules and their uses
US7521063B2 (en) 2000-01-14 2009-04-21 The United States Of America As Represented By The Department Of Health And Human Services Multiple CPG oligodeoxynucleotides and their use to induce an immune response
US20030144229A1 (en) * 2000-01-14 2003-07-31 The Government Of The U.S.A., The Secretary Of The Department Of Health And Human Services Multiple CpG oligodeoxynucleotides and their use to induce an immune response
US7378275B2 (en) 2000-04-07 2008-05-27 Heska Corporation Canine IL-13 receptor alpha-1 subunit nucleic acid molecules
US20080287665A1 (en) * 2000-04-07 2008-11-20 Mccall Catherine A CANINE IgG NUCLEIC ACID MOLECULES
US20040142372A1 (en) * 2000-04-07 2004-07-22 Mccall Catherine A. Compositions and methods related to canine IgG and canine IL-13 receptors
US6703360B2 (en) 2000-04-07 2004-03-09 Heska Corporation Compositions and methods related to canine IgG and canine IL-13 receptors
US20030194391A1 (en) * 2001-03-03 2003-10-16 Claire Ashman Vaccine
US20050186209A1 (en) * 2001-03-03 2005-08-25 Claire Ashman Vaccine
US20050214355A1 (en) * 2001-07-27 2005-09-29 Klinman Dennis M Washing and chilling apparatus and method
US7666674B2 (en) 2001-07-27 2010-02-23 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Use of sterically stabilized cationic liposomes to efficiently deliver CPG oligonucleotides in vivo
US7959934B2 (en) 2001-08-14 2011-06-14 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Method for rapid generation of mature dendritic cells
US7354909B2 (en) 2001-08-14 2008-04-08 The United States Of America As Represented By Secretary Of The Department Of Health And Human Services Method for rapid generation of mature dendritic cells
US20080241176A1 (en) * 2001-08-14 2008-10-02 The Gov. Of The U.S.A As Represented By The Secretary Of The Dept. Of Health & Human Services Method for rapid generation of mature dendritic cells
US20050002918A1 (en) * 2001-11-09 2005-01-06 Neopharm, Inc. Selective treatment of IL-13 expressing tumors
US20050142105A1 (en) * 2001-12-04 2005-06-30 Puri Raj K. Chimeric molecule for the treatment of th2-like cytokine mediated disorders
US7541040B2 (en) * 2001-12-04 2009-06-02 The United States Of America As Represented By The Department Of Health And Human Serivces Chimeric molecule for the treatment of th2-like cytokine mediated disorders
US7615227B2 (en) 2001-12-20 2009-11-10 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Use of CpG oligodeoxynucleotides to induce angiogenesis
US8466116B2 (en) 2001-12-20 2013-06-18 The Unites States Of America As Represented By The Secretary Of The Department Of Health And Human Services Use of CpG oligodeoxynucleotides to induce epithelial cell growth
US20050026245A1 (en) * 2001-12-20 2005-02-03 Klinman Dennis M. Use of cpg oligodeoxynucleotides to induce angiogenesis
US7935351B2 (en) 2001-12-20 2011-05-03 The United States Of America As Represented By The Department Of Health And Human Services Use of CPG oligodeoxynucleotides to induce angiogenesis
US20110020363A1 (en) * 2002-03-22 2011-01-27 Zenyth Operations Pty Ltd. Monoclonal antibody against interleukin-13 receptor alpha 1 (il-13ralpha1)
US7785590B2 (en) 2002-03-22 2010-08-31 Zenyth Operations Pty Ltd. Monoclonal antibody against interleukin-13 receptor alpha 1 (IL-13Ra1)
US20050058645A1 (en) * 2002-03-22 2005-03-17 Amrad Operations Pty. Ltd. Monoclonal antibody against Interleukin-13 receptor alpha 1 (IL-13Ralpha1)
US8221755B2 (en) 2002-03-22 2012-07-17 Zenyth Operations Pty Ltd. Monoclonal antibody against interleukin-13 receptor alpha 1 (IL-13Ralpha1)
US20040105872A1 (en) * 2002-09-18 2004-06-03 The Government Of The Usa As Represented By The Secretary Of The Dept. Of Health & Human Services Method of treating and preventing infections in immunocompromised subjects with immunostimulatory CpG oligonucleotides
US8263091B2 (en) 2002-09-18 2012-09-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Method of treating and preventing infections in immunocompromised subjects with immunostimulatory CpG oligonucleotides
US20060099652A1 (en) * 2003-03-26 2006-05-11 Neopharm, Inc. IL 13 receptor alpha 2 antibody and methods of use
US20060140948A1 (en) * 2004-11-17 2006-06-29 Ian Foltz Fully human monoclonal antibodies to IL-13
US7585500B2 (en) 2004-11-17 2009-09-08 Amgen Inc. Fully human monoclonal antibodies to IL-13
US7994302B2 (en) 2004-11-17 2011-08-09 Amgen Inc. Fully human monoclonal antibodies to IL-13
WO2006113614A2 (en) 2005-04-15 2006-10-26 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Service National Institutes Of Health Methods of treating and preventing inflammatory bowel disease involving il-13 and nkt cells
US8343461B2 (en) 2006-03-08 2013-01-01 Wake Forest University Health Sciences Molecular signature of cancer
EP2946786A1 (en) 2006-03-08 2015-11-25 Wake Forest University Health Sciences Soluble monomeric Ephrin A1
US11207380B2 (en) 2006-03-08 2021-12-28 Wake Forest University Health Sciences Molecular signature of cancer
US9974830B2 (en) 2006-03-08 2018-05-22 Wake Forest University Health Sciences Molecular signature of cancer
US9290558B2 (en) 2006-03-08 2016-03-22 Wake Forest University Health Sciences Molecular signature of cancer
US20090285799A1 (en) * 2006-10-19 2009-11-19 Andrew Donald Nash Antibody Antagonists of Interleukin-13 Receptor Alpha1
US8207304B2 (en) 2006-10-19 2012-06-26 Csl Limited Antibody antagonists of interleukin-13 receptor α1
WO2008049098A3 (en) * 2006-10-19 2009-04-09 Merck & Co Inc ANTIBODY ANTAGONISTS OF INTERLEUKIN-13 RECEPTOR α1
US9296785B2 (en) 2009-04-17 2016-03-29 Wake Forest University Health Sciences IL-13 receptor binding peptides
US9878013B2 (en) 2009-04-17 2018-01-30 Wake Forest University Health Sciences IL-13 receptor binding peptides
US9005600B2 (en) 2010-04-16 2015-04-14 Wake Forest University Health Sciences Compounds useful for multi-level specific targeting of cancer cells
US11136367B2 (en) 2010-04-16 2021-10-05 Wake Forest University Health Sciences Multi-level specific targeting of cancer cells
US9771404B2 (en) 2010-04-16 2017-09-26 Wake Forest University Health Sciences Multi-level specific targeting of cancer cells
US8362207B2 (en) 2010-04-16 2013-01-29 Wake Forest University Health Sciences Multi-level specific targeting of cancer cells with IL-13
US10233224B2 (en) 2010-04-16 2019-03-19 Wake Forest University Health Sciences Nucleic acids encoding a fusion protein useful for multi-level specific targeting of cancer cells
US10519210B2 (en) 2010-04-16 2019-12-31 Wake Forest University Health Sciences Multi-level specific targeting of cancer cells
WO2015070210A1 (en) 2013-11-11 2015-05-14 Wake Forest University Health Sciences Epha3 and multi-valent targeting of tumors
US10752667B2 (en) 2013-11-11 2020-08-25 Wake Forest University Health Sciences EPHA3 and multi-valent targeting of tumors
US9975942B2 (en) 2013-11-11 2018-05-22 Wake Forest University Health Services EPHA3 And multi-valent targeting of tumors
US11697675B2 (en) 2013-11-11 2023-07-11 Wake Forest University Health Sciences EPHA3 and multi-valent targeting of tumors
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria

Also Published As

Publication number Publication date
JP4202417B2 (en) 2008-12-24
EP1007696B1 (en) 2008-12-17
AU5311096A (en) 1996-10-08
CA2215122A1 (en) 1996-09-26
AU714541B2 (en) 2000-01-06
JP2000511042A (en) 2000-08-29
DE69637781D1 (en) 2009-01-29
EP1007696A1 (en) 2000-06-14
ES2319827T3 (en) 2009-05-12
WO1996029417A1 (en) 1996-09-26
US5614191A (en) 1997-03-25
ATE417931T1 (en) 2009-01-15

Similar Documents

Publication Publication Date Title
US5919456A (en) IL-13 receptor specific chimeric proteins
US6518061B1 (en) IL-13 receptor specific chimeric proteins and uses thereof
US6428788B1 (en) Compositions and methods for specifically targeting tumors
US6296843B1 (en) Mutagenized IL 13-based chimeric molecules
CA2187283C (en) Circularly permuted ligands and circularly permuted chimeric molecules
US5608039A (en) Single chain B3 antibody fusion proteins and their uses
US5889157A (en) Humanized B3 antibody fragments, fusion proteins, and uses thereof
US6576232B1 (en) IL13 mutants
AU2001288405B2 (en) Amino acid substitution mutants of interleukin 13
US6884603B2 (en) Nucleic acids encoding IL13 mutants
AU7863900A (en) Il13 mutants
US6630576B2 (en) Amino acid substitution mutants of interleukin 13
US20030129132A1 (en) IL-13 receptor specific chimeric proteins &amp; uses thereof
US20040136959A1 (en) Sensitization of cancer cells to immunoconjugate-induced cell death by transfection with il -13 receptor alpha chain
WO2002017968A2 (en) Sensitization of cancer cells to immunoconjugate-induced cell death by transfection with il-13 receptor alpha chain-2
AU2001288405A1 (en) Amino acid substitution mutants of interleukin 13

Legal Events

Date Code Title Description
STCF Information on status: patent grant

Free format text: PATENTED CASE

FPAY Fee payment

Year of fee payment: 4

FPAY Fee payment

Year of fee payment: 8

FPAY Fee payment

Year of fee payment: 12